CA2903595A1 - Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use - Google Patents

Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use Download PDF

Info

Publication number
CA2903595A1
CA2903595A1 CA2903595A CA2903595A CA2903595A1 CA 2903595 A1 CA2903595 A1 CA 2903595A1 CA 2903595 A CA2903595 A CA 2903595A CA 2903595 A CA2903595 A CA 2903595A CA 2903595 A1 CA2903595 A1 CA 2903595A1
Authority
CA
Canada
Prior art keywords
seq
light chain
fap
heavy chain
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA2903595A
Other languages
French (fr)
Other versions
CA2903595C (en
Inventor
Peter Bruenker
Sherif Daouti
Claudia Ferrara Koller
Guy Georges
Sandra GRAU-RICHARDS
Ralf Hosse
Christian Klein
Maximiliane KOENIG
Joerg Moelleken
Ekkehard Moessner
Huifeng Niu
Kathryn E. Packman
Valeria RUNZA
Stefan Seeber
Pablo Umana
Inja Waldhauer
Huisheng Wang
Barbara Weiser
Ningping FENG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Roche Glycart AG
Original Assignee
Roche Glycart AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roche Glycart AG filed Critical Roche Glycart AG
Publication of CA2903595A1 publication Critical patent/CA2903595A1/en
Application granted granted Critical
Publication of CA2903595C publication Critical patent/CA2903595C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/624Disulfide-stabilized antibody (dsFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/66Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a swap of domains, e.g. CH3-CH2, VH-CL or VL-CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present disclosure relates to bispecific antibodies comprising at least one antigen binding site specific for DR5 and at least one antigen binding site specific for FAP, antibodies specific for DR5, methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.

Description

DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter le Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS
THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME:
NOTE POUR LE TOME / VOLUME NOTE:

BISPECIFIC ANTIBODIES SPECIFIC FOR FAP AND DR5, ANTIBODIES SPECIFIC

FIELD OF THE INVENTION
The present invention relates to bispecific antibodies comprising a first antigen binding site specific for Death Receptor 5 (DR5) and a second antigen binding site specific for Fibroblast Activation Protein (FAP), antibodies specific for DR5, methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
BACKGROUND
Monoclonal antibodies are powerful therapeutic agents in the treatment of cancer since they selectively target antigens which are differentially expressed on cancer cells. Targeting of the TRAIL (TNF related apoptosis inducing ligand) death receptors on cancer cells with agonistic monoclonal antibodies represents a new generation of monoclonal antibody therapy, as they are able to directly induce apoptosis of targeted cells.
Upon binding of TRAIL, death receptors of the TNFR-SF family such as DR4 and become trimerized. The trimerization induces the extrinsic apoptotic pathway and a complex cascade of events including Caspase activation, which finally result in the killing of the target cells. Apoptosis induction is further enhanced if hyperclustering of DR5 (i.e.
the clustering of multiple trimers) takes place. Although death receptors are widely expressed on a variety of cell types, induction of apoptosis via the extrinsic pathway is restricted to tumor cells. Since agonistic DR4 or DRS binding antibodies are able to cross-link death receptors and hence induce apoptosis, these receptors are interesting targets in cancer therapy. At least eight death receptor targeting molecules entered clinical development and have been assessed in clinical trials for possible treatment of different indications such as advanced solid tumors like colorectal or lung cancers. In addition there have been attempts to treat other indications such as lymphoma and /
or multiple myeloma.
Drozitumab, a fully human DRS agonistic antibody described in US2007 / 0031414 Al and W02006/083971, shows some in vitro apoptotic activity in the absence of cross-linking at high concentrations. However, in vivo data revealed a different mode of action: In FcyR mutant mice (or when antibody variants were used in which FcyR binding was inhibited) Drozitumab was inactive indicating that the in vivo activity of this molecule is mainly dependent on FcyR
-2-mediated cross-linking. This molecule was tested up to clinical phase II, seemed to be save (no MTD up to 20 mg/kg was reached) but did not demonstrate any significant efficacy.
Conatumumab (described in EP1922337A), is another fully human DR5 agonistic antibody. The activity of Conatumumab is strictly dependent on cross-linking via Fc receptors.
In contrast to Drozitumab this antibody is non-ligand blocking. Also this molecule only showed very limited efficacy in clinical trials.
LBY-135, a chimeric DRS antibody, exhibits similar characteristics as Conatumumab with respect to cross-linking dependent activity and non-ligand blocking property and did not demonstrate any significant efficacy in monotherapy. In addition, LBY-135 showed signs of immunogenicity in part of the enrolled patients of a phase I trial.
Dulanermin, a recombinantly produced natural ligand of DR4 and DR5 (TRAIL), only showed limited objective responses in clinical trials. The use of the natural ligand has somehow disadvantageous: TRAIL targets multiple receptors including both the death receptors and decoy receptors and, therefore, selectivity is a concern. In addition, TRAIL has a much shorter blood half-life compared with monoclonal anti- DR antibodies, a factor which affects dose and schedule parameters. The very short blood half-life of TRAIL requires large and frequent doses compared with monoclonal anti-DR antibodies. In addition recombinant TRAIL is very difficult and tedious to produce.
The development of all three DRS agonistic antibodies and the ligand described above was discontinued.
Two additional fully human antibodies, Mapatumumab (anti DR4) and Lexatumumab (anti DRS) are still in development although also these molecules did not exhibit promising efficacy in monotherapy.
Tigatuzumab is a humanized DRS agonistic antibody which is described as being active in vitro (already at low concentrations) in the absence of secondary cross-linking which of course bears the risk of systemic toxicity issues. However, as all the other described agonistic DRS
antibodies, also this molecule has not demonstrated convincing efficacy in Ph I / Ph II studies so far and the maximally tolerated dose MTD only was demonstrated up to 8 mg/kg.
A different approach to induce apoptosis by targeting a death receptor is pursued with the molecule TAS266, a tetrameric DRS binding nanobody (W02011098520A1). Due to the tetravalent configuration of DRS binding moieties, it is thought that DRS
cross-linking is increased compared to standard bivalent antibodies, which may result in increased activity.
However, due to their small size, these molecules have the disadvantage of a rather short half-life
-3-(compared to antibodies). In addition there is an increased risk of systemic toxicity since this tetrameric molecule is not targeted to the tumor.
Combining the DR5 antibody Drozitumab with a tumor antigen binding moiety or an antigen present in the stroma surrounding the tumor in a bispecific antibody platform has been described by the inventors of the present application as a new approach to achieve two effects:
firstly the DRS binding antibody can be targeted to the tumor site which could avoid potential systemic toxicity issues (especially when using a DRS antibody exhibiting cross-linking independent activity). Secondly, this tumor or tumor stroma targeting moiety then also serves as the cross-linking unit to induce DRS hyperclustering and subsequently tumor site specific apoptosis. The basic concept has been demonstrated using Drozitumab_scFv fusion molecules targeting different tumor types (see WO 2011/039126).
Of particular interest therein were bispecific antibodies binding DRS and Human Fibroblast Activation Protein (FAP; GenBank Accession Number AAC51668). Human FAP was originally identified in cultured fibroblasts using the monoclonal antibody (mAb) F19 (described in WO 93/05804, ATCC Number HB 8269). Homologues of the protein were found in several species, including mice (Niedermeyer et al., Int J Cancer 71, 383-389 (1997), Niedermeyer et al., Eur J Biochem 254, 650-654 (1998); GenBank Accession Number AAH19190). FAP has a unique tissue distribution: its expression was found to be highly upregulated on reactive stromal fibroblasts of more than 90% of all primary and metastatic epithelial tumors, including lung, colorectal, bladder, ovarian and breast carcinomas, while it is generally absent from normal adult tissues (Rettig et al., Proc Natl Acad Sci USA 85, 3110-3114 (1988); Garin-Chesa et al., Proc Natl Acad Sci USA 87, 7235-7239 (1990)). Subsequent reports showed that FAP is not only expressed in stromal fibroblasts but also in some types of malignant cells of epithelial origin, and that FAP expression directly correlates with the malignant phenotype (Jin et al., Anticancer Res 23, 3195-3198 (2003)). Surprisingly the inventors found that a bispecific antibody targeting FAP
in the stroma and DRS on the tumor cell actually induces apoptosis despite the targets being situated on different cells.
Upon further investigation the inventors of the present application found that the scFv containing bispecific molecules described in W02011/039126 all have some intrinsic issues with respect to productivity, stability and aggregate formation leading to suboptimal, non-specific activity.
In the present application, novel bispecific antibodies targeting FAP and DRS
are provided.
The inventors of the present application developed novel DRS binding moieties that are only active after crosslinking. Hence induction of tumor cell apoptosis is dependent on DRS
hypercrosslinking via FAP and is independent on Fc/ FcR interactions.
Therefore in addition to
-4-bispecific antibodies specific for FAP and DR5, also novel antibodies binding to DRS are provided therein.
In contrast, the activity of conventional DRS targeting molecules as described above is dependent on Fc Receptor (FcR) mediated hyperclustering, and is influenced by the immune infiltration and activation status in the tumor (Li and Ravetch, PNAS 2012;
Wilson, Cancer Cell 2011; W02011098520A1). The Fc/FcR interactions can be impaired by physiological human IgG levels. Thus the activity of conventional DRS targeting molecules is often limited to a few infiltrating cells (Moessner, Blood 2010). By using a bispecific antibody targeting both DRS and FAP, the percentage of sensitive tumor cells can be significantly increased by hypercrosslinking via FAP and the risk of an intrinsic resistance to DRS agonists is decreased.
The novel DRS
binding moieties are only active after crosslinking with FAP, which could result in an improved safety and toxicology profile compared to the DRS binders Apomab and Tigatuzumab. The DRS
agonists that have been tested so far were safe in the clinic, however, these clinical programs have been impeded by a low efficacy of the DRS targeting molecules.
In addition, the preferred novel DRS binding moieties bind to a different epitope than Drozitumab.
Importantly the novel DRS binding moieties can be employed in many bispecific DRS-FAP targeting antibody formats, including both novel and established bispecific formats. In contrast, only C-terminal fusions of a FAP binding moiety are possible with Drozitumab-based bispecific DRS-FAP targeting antibody formats, as any N-terminal fusion to Drozitumab results in inactive molecules. The provision of the new DRS binding moieties thus significantly expands the possibilities of employing the DRS targeting moiety in various bispecific DRS-FAP targeting antibody formats. This is particularly important as some bispecific antibody formats have superior characteristics in terms of producability and activity.
Provided therein are novel bispecific antibodies comprising novel DRS binding moieties and a affinity matured FAP binding moiety.
The bispecific antibodies of the present invention are provided in a bispecific antibody format, wherein one or more crossover- Fab fragments are fused to an IgG
molecule. Crossover Fab fragments are Fab fragments wherein either the variable regions or the constant regions of the heavy and light chain are exchanged. Bispecific antibody formats comprising crossover Fab fragments have been described, for example, in W02009080252, W02009080253, W02009080251, W02009080254, W02010/136172, W02010/145792 and W02013/026831.
So far, only DRS-FAP bispecific antibodies in a scFv containing format have been described (W02011/039126). In addition to the advantageous properties of the novel DRS
5 PCT/EP2014/056511 binders disclosed therein, use of a crossover-Fab based bispecific format results in improved yield and less aggregates and side-products during production. In addition, scFv- and scFab-based DR5-FAP targeting bispecific antibodies demonstrated the tendency to form aggregates and therefore bear a higher riskto non-specifically crosslinkDR5 even in absence of FAP. Hence these formats have the disadvantage of potentially inducing apoptosis in non-target cells (i.e.
healthy cells).
The DRS and FAP binding moieties of the novel bispecific antibodies provided herein exhibit superior in vivo efficacy compared to conventional DRS antibodies. The preferred bispecific antibodies of the present invention bind with a high affinity to FAP on the tumor stroma and with a lower affinity to DRS on the tumor cell. Moreover, the DRS
and FAP targeting bispecific antibodies provided herein do not crossreact with closely related proteins DR4, DcR1, DcR2 (closely related to DRS) and DPPIV (closely related to FAP). In addition it is now for the first time possible to provide a DRS-FAP bispecific antibody in various bispecific formats with no limitation as to the number of valencies of each binding specifity.
To summarize, the novel bispecific DRS-FAP antibodies provided therein are highly specific and potent: They selectively induce apoptosis by DRS hyperclustering in tumor cells in a FAP dependent manner, with low binding to normal cells.
SUMMARY
The present invention relates to bispecific antibodies combining a Death Receptor 5 (DRS) targeting antigen binding site with a second antigen binding site that targets Fibroblast Activation Protein (FAP). By that the death receptors become cross linked and apoptosis of the targeted tumor cell is induced. The advantage of these bispecific death receptor agonistic antibodies over conventional death receptor targeting antibodies is the specificity of induction of apoptosis only at the site where FAP is expressed as well as the higher potency of these bispecific antibodies due to the induction of DRS hyperclustering. In addition, novel antibodies binding to DRS are provided. As outlined above the inventors of the present invention developed novel DRS binding moieties with superior properties compared to known DRS
binders that are incorporated into novel and advantageous DRS-FAP bispecific antibodies.
In one embodiment, the invention provides a bispecific antibody that binds to death receptor 5 (DRS) and Fibroblast Activation Protein (FAP), comprising at least one antigen binding site specific for DRS, comprising
-6-(a) a heavy chain CDR1 consisting of SEQ ID NO.:1, SEQ ID NO.:17 and SEQ ID
NO.:75;
(b) a heavy chain CDR2 of SEQ ID NO.:2, SEQ ID NO.:18, SEQ ID NO.:25 and SEQ
ID
NO.:83;
(c) a heavy chain CDR3 of SEQ ID NO. :3, SEQ ID NO.:19, SEQ ID NO. :84, SEQ ID
NO. :96, SEQ ID NO. :98, SEQ ID NO.:104 and SEQ ID NO.:108;
(d) a light chain CDR1 of SEQ ID NO. :4, SEQ ID NO. :20, SEQ ID NO. :27 and SEQ ID
NO.:86;
(e) a light chain CDR2 of SEQ ID NO. :5, SEQ ID NO. :21 and SEQ ID NO. :28;
and (f) a light chain CDR3 of SEQ ID NO. :6, SEQ ID NO. :22, SEQ ID NO. :87, SEQ
ID
NO. :99, SEQ ID NO.:105, SEQ ID NO.:109 and SEQ ID NO. :97;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO.:9 and SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:10 and SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO.:11 and SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO.:12 and SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO.:13 and SEQ ID NO.:37;
(f) a light chain CDR3 of SEQ ID NO.:14 and SEQ ID NO. :38.
In one embodiment, the invention provides a bispecific antibody wherein the antigen binding site specific for DR5 comprises (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO. :5;
(f) a light chain CDR3 of SEQ ID NO.:6 and the antigen binding site specific for FAP comprises (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
-7-(f) a light chain CDR3 of SEQ ID NO.:14.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5 comprising a variable heavy chain and a variable light chain comprising an amino acid sequence of: SEQ ID NO. :7 and SEQ ID NO. :8; SEQ ID NO.:23 and SEQ ID
NO. :24; SEQ ID NO.:26 and SEQ ID NO. :24; SEQ ID NO.:23 and SEQ ID NO. :29;
SEQ ID
NO.:23 and SEQ ID NO.:30; SEQ ID NO.:26 and SEQ ID NO.:31; SEQ ID NO.:26 and SEQ
ID NO.:32; SEQ ID NO.:26 and SEQ ID NO.:30; SEQ ID NO.:23 and SEQ ID NO.:31;
SEQ
ID NO.:82 and SEQ ID NO.:85; SEQ ID NO.:100 and SEQ ID NO.:101; SEQ ID NO.:102 and SEQ ID NO.:103; SEQ ID NO.:106 and SEQ ID NO.:107; SEQ ID NO. :94 and SEQ
ID NO.:
95;
and at least one antigen binding site specific for FAP comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO.:15 and SEQ ID NO. :39 ; and a variable light chain comprising an amino acid sequence of SEQ ID NO.:16 and SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5 comprising a variable heavy chain comprising an amino acid sequence of SEQ ID
NO.:7 and a variable light chain comprising an amino acid sequence of SEQ ID
NO. :8; and at least one antigen binding site specific for FAP comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
Preferably said bispecific antibody is human or humanized.
In one embodiment, the bispecific antibody comprises an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DR5, and at least one Fab fragment comprising the antigen binding site specific for FAP.
In one embodiment, the bispecific antibody comprises an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DRS, and at least one Fab fragment comprising the antigen binding site specific for FAP, wherein at least one of the Fab fragments is connected to the first or second subunit of the Fc domain via the light chain (VLCL) and at least one Fab
-8-fragment is connected to the first or second subunit of the Fc domain via the heavy chain (VHCH1).
In one embodiment, the bispecific antibody comprises a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DR5, wherein said Fab fragments are connected at the C-terminus of the constant light chain (CL) to the first or second subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain.
In one embodiment, the bispecific antibody comprises a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DRS, wherein said Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the C-terminus of the constant light chain (CL) to the first or second subunit of the Fc domain.
In one embodiment, the bispecific antibody comprises an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DRS, and at least one Fab fragment comprising the antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In one embodiment, the bispecific antibody comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In one embodiment said bispecific antibody is bivalent both for DRS and FAP.
-9-In one embodiment, the bispecific antibody comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DR5, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In one embodiment said bispecific antibody is bivalent for DRS and monovalent for FAP.
In one embodiment, the bispecific antibody comprises an Fc domain, three Fab fragments comprising each an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In one embodiment said bispecific antibody is trivalent for DRS and monovalent for FAP.
In one embodiment, the bispecific antibody comprises an Fc domain, one Fab fragment comprising an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In one embodiment said bispecific antibody is monovalent for DRS and monovalent for FAP.
In one embodiment said bispecific antibody comprises an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DRS, and at least one Fab fragment comprising the antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of the Fab fragment(s) comprising an antigen binding site specific for FAP are exchanged.
In one embodiment at least one of said Fab fragments is connected to the Fc domain via a peptide linker.
In one embodiment said bispecific antibody comprises an Fc domain, which comprises one or more amino acid substitution that reduces binding to Fc receptors and/or effector function. In one embodiment said one or more amino acid substitution is at one or more positions selected from the group of L234, L235, and P329. In one embodiment each subunit of the Fc domain
-10-comprises three amino acid substitutions that abolish binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G.
In one embodiment a bispecific antibody is provided wherein the Fc part of the first heavy chain comprises a first dimerization module and the Fc part of the second heavy chain comprises a second dimerization module allowing a heterodimerization of the two heavy chains of the antibody. In one embodiment the first dimerization module comprises knobs and the second dimerization module comprises holes according to the knobs into holes strategy.
In a further embodiment an antibody that specifically binds to DRS is provided, comprising (a) a heavy chain complementarity determining region 1 (CDR1) selected from the group consisting of SEQ ID NO.:1, SEQ ID NO.:17 and SEQ ID NO.:75;
(b) a heavy chain complementarity determining region 2(CDR2) selected from the group of SEQ ID NO. :2, SEQ ID NO.:18, SEQ ID NO.:25 and SEQ ID NO.:83;
(c) a heavy chain complementarity determining region 3 (CDR3) selected from the group of SEQ ID NO. :3, SEQ ID NO.:19, SEQ ID NO.:84, SEQ ID NO.:96, SEQ ID NO.:98, SEQ ID
NO.:104 and SEQ ID NO.:108;
(d) a light chain CDR1 selected from the group of SEQ ID NO. :4, SEQ ID NO.
:20, SEQ
ID NO. :27 and SEQ ID NO. :86;
(e) a light chain CDR2 selected from the group of SEQ ID NO. :5, SEQ ID NO.
:21 and SEQ ID NO. :28; and (f) a light chain CDR3 selected from the group of SEQ ID NO.:6, SEQ ID NO.
:22, SEQ ID
NO. :87, SEQ ID NO. :99, SEQ ID NO.:105, SEQ ID NO.:109 and SEQ ID NO. :97;
In a further embodiment an antibody that specifically binds to DRS is provided, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO. :5;
(f) a light chain CDR3 of SEQ ID NO.:6
-11 -In a further embodiment an antibody that specifically binds to DR5 is provided, comprising a variable heavy chain and a variable light chain comprising an amino acid sequence selected from the group of: SEQ ID NO. :7 and SEQ ID NO. :8; SEQ ID NO. :23 and SEQ ID NO.
:24; SEQ
ID NO.:26 and SEQ ID NO. :24; SEQ ID NO. :23 and SEQ ID NO. :29; SEQ ID NO.
:23 and SEQ ID NO.:30; SEQ ID NO.:26 and SEQ ID NO.:31; SEQ ID NO.:26 and SEQ ID
NO.:32;
SEQ ID NO.:26 and SEQ ID NO.:30; SEQ ID NO.:23 and SEQ ID NO.:31; SEQ ID
NO.:82 and SEQ ID NO.:85; SEQ ID NO.:100 and SEQ ID NO.:101; SEQ ID NO.:102 and SEQ
ID
NO.:103; SEQ ID NO.:106 and SEQ ID NO.:107; SEQ ID NO. :94 and SEQ ID NO. :95;
In a further embodiment an antibody that specifically binds to DR5 is provided comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :7 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:8.
In a second object the present invention relates to a pharmaceutical composition comprising a bispecific antibody or the antibody that specifically binds to DR5 of the present invention.
In a third object the present invention relates to a bispecific antibody targeting DRS and FAP or an antibody that specifically binds to DRS of the present invention for the treatment of cancer. In one preferred embodiment said cancer is pancreatic cancer or colorectal carcinoma. In another embodiment, use of the bispecific antibody or an antibody that specifically binds to DRS
as a medicament is provided. Preferably said use is for the treatment of cancer, preferably pancreatic cancer or colorectal carcinoma.
In further objects the present invention relates to a nucleic acid sequence comprising a sequence encoding a heavy chain of a bispecific antibody or an antibody that specifically binds to DRS of the present invention, a nucleic acid sequence comprising a sequence encoding a light chain of a bispecific antibody or an antibody that specifically binds to DRS
of the present invention, an expression vector comprising a nucleic acid sequence of the present invention and to a prokaryotic or eukaryotic host cell comprising a vector of the present invention. In addition a method of producing an antibody comprising culturing the host cell so that the antibody is produced is provided.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1: FACS binding analysis of two different cell lines (5W872 and GM05389) for expression levels of human fibroblast activation protein (FAP) (A). The fluorescence intensity
-12-measured with different concentrations of an anti FAP antibody is shown over a range of three magnitudes (black, grey and hatched bars). Negative control reactions as secondary antibody and cells only are shown as stippled and white bars, respectively. While the GM05389 cells demonstrate expression of FAP over all tested antibody concentrations that was above background, with the SW872 cells FAP expression only could be detected with the highest antibody concentration used (10 p g / ml), indicating that these cells are not suitable for FAP
based binding / apoptosis induction experiments. In addition it is shown that this cell line hardly undergoes Drozitumab mediated apoptosis (B). Drozitumab alone or another, commercially available anti DRS antibody did not induce relevant DNA fragmentation. Only when Drozitumab is cross-linked with an anti-human Fc antibody a detectable low level apoptosis induction can be observed.
Figure 2: Detection of DRS expression on two different human tumor cell lines (breast cancer cell line MDA-MB-231 and the renal carcinoma cell line ACHN) via FACS binding with Drozitumab and subsequent detection with a labeled anti Fc antibody. Both cell lines show comparable, low expression levels of human DRS.
Figure 3: DNA fragmentation ELISA assay for detection of apoptosis. ACHN (A) or MDA-MB-231 (B) target cells were either cultivated alone or in presence of an equal number of FAP
expressing GM05389 fibroblasts. DRS-FAP bispecific antibodies (scFv fusions) were added at a concentration of 0.1 p g / ml (A) and 0.7 nM , respectively (B) and cells were incubated for 24 h prior to detection of DNA fragmentation. For cross-linking of Drozitumab, 0.1 ng / ml secondary anti Fc antibody was used.
Figure 4: DNA fragmentation ELISA assay for detection of apoptosis. Apoptosis induction on ACHN cells by bispecific DRS-FAP antibodies (Drozitumab with fused scFab) in the presence or absence of FAP expressing fibroblasts (GM05389). Activity of bispecific molecules with different FAP binding moieties are compared to Drozitumab with or without cross-linking by a secondary anti Fc antibody. Antibodies were used in two different concentrations (1.0 p g/ml and 0.1 p g/m1). In these settings bispecific molecules containing the 4G8 scFab fusion (B) showed superior apoptosis induction activity in a DNA fragmentation assay compared to the 3F2 scFab containing molecules (A).
Figure 5: Schematic representation of two different bispecific DRS-FAP
molecules in which the anti FAP CrossFab moiety by a standard (G45)4 linker is fused to the C-terminus of Drozitumab IgG. In one case (A) the VHCL CrossFab is fused to Drozitumab IgG while in (B) the VLCH1 chain is connected to the IgG as indicated. These two molecules are only examples for possible formats using IgG-CrossFab combinations which also were used for the bispecific molecules containing newly isolated DRS binders. Other possibilities include the crossing in the IgG part of the molecule, the implementation of salt bridges and charged residues to stabilize the CrossFabs or the use of different linker lengths and sequences.
Figure 6: Comparison of aggregate contents during purification of different bispecific DRS-FAP
antibody formats. All formats consist of Drozitumab IgG with a FAP binding domain (clone 4G8) fused to the C-terminus. FAP binding moieties consist of either a disulfide stabilized scFv (A), a
-13-scFab (B) or a CrossFab (C). The chromatograms of the preparative size exclusion chromatography showed clear differences between the three constructs. While the production of the scFv and CrossFab molecules was comparable, the scFab containing construct showed lower yield and higher aggregate content. From this comparison the CrossFab containing molecule looked most promising.
Figure 7: Analysis of FACS binding on recombinant, human FAP expression HEK293 cells.
DR5-FAP bispecific molecules in the new CrossFab format were compared to the scFab molecule, the parental 4G8 IgG and an unrelated negative control, Drozitumab.
The two CrossFab molecules were shown to bind to FAP at least in the same range as the IgG control.
Figure 8: Surface plasmon resonance (SPR, Biacore) analysis of simultaneous binding of bispecific Drozitumab-FAP molecules (CrossFabs) to recombinant human DRS and FAP.
Biotinylated human DR5-Fc as the ligand was immobilized onto a streptavidin chip followed by injection of the first analyte (bispecific CrossFab molecules). After binding to DR5-Fc (90 sec association) and a short dissociation period (10 sec), recombinant soluble FAP
(human or murine) in different concentrations (100 nM, 500 nM) was added as the second analyte and the additional response was measured. Binding of Drozitumab-X-FAP_A format (A and B) was compared to Drozitumab-X-FAP_B (C and D). Each construct was measured for binding DRS and human (A, C) or murine FAP (B, D). By this analysis for all tested molecules a simultaneous binding to DRS and human / murine FAP could be demonstrated.
Figure 9: DNA fragmentation ELISA assay for detection of apoptosis. Results of an apoptosis induction experiment in which two CrossFab molecules (Drozitumab-X-Fab-A and Drozitumab-X-Fab-B) were compared to Drozitumab and hyper-cross-linked Drozitumab in a two cell line (MDA-MB-231 and GM05389) co-culture assay. (A) Induction of apoptosis was detected after 24 hrs using a cell death detection ELISA. All tested constructs were used in concentrations of 7 nM and 0.7 nM. Apoptosis induction of target and effector cells is compared to apoptosis of target cells alone. (B) Comparison of co-culture bystander apoptosis induction in Cell Death Detection ELISA (DNA fragmentation) with Drozitumab (+/- Fc cross-linking) vs.
bispecific DRS-FAP molecules consisting of a FAP CrossFab fused to the C-terminus of Drozitumab heavy chain. The molecule containing the affinity matured FAP binding moiety (28H1) shows superior activity compared to the molecule containing the lower affinity FAP binder.
Figure 10: DNA fragmentation ELISA assay for detection of apoptosis. Induction of apoptosis via bispecific Drozitumab-FAP CrossFab molecules is dependent on cross-linking via FAP.
Figure 10 shows the results of apoptosis induction on MDA-MB-231 cells by CrossFab molecules cross-linked by recombinant human FAP coated to an ELISA plate.
While the control molecule (Drozitumab cross-linked via anti Fc antibody) shows similar apoptosis induction, independent on coating of FAP or an unrelated control protein, the bispecific constructs only exhibited significant apoptosis activity when FAP was coated. With coating of the control plasmid only at highest construct concentration (7 nM) apoptosis could be detected which probably is due to the basal FAP expression of the MDA-MB-231 cell line.
Apoptosis activity was similar for both tested CrossFab molecules and in the same range as observed with hyper-cross-linked Drozitumab.
-14-Figure 11: Comparison of apoptosis induction (DNA fragmentation) on different human tumor cell lines in a co-culture experiment with FAP expressing human fibroblasts GM05389.
Drozitumab and hyper-cross-linked Drozitumab is compared to a bispecific Drozitumab-4G8 CrossFab molecule (all at concentrations of 7.0 and 0.7 nM). Used tumor cell lines were MDA-MB-231 (breast cancer), U-87MG (glioblastoma), FaDu (squamous carcinoma) or A549 (lung carcinoma). Apoptosis induction at a concentration of 7 nM was similar for all four cell lines while at 0.7 nM a more pronounced difference between the cell lines was observed.
Figure 12: Schematic representation of the human DR5 antigen constructs used for isolation, screening and characterization of novel DR5 binders. For all constructs the same DRS domain (extracellular domain, ECD; aa 56 ¨ 207) was used either alone (A) as dimeric Fc fusion (B) or as monomeric Fc fusion (C) using the knob-into-hole technology. All antigens were transiently transfected and produced in HEK293 EBNA cells.
Figure 13: Screening of 46 unique DRS binders (isolated by phage display) for induction of apoptosis on MDA-MB-231 cells (DNA fragmentation assay) after hyper-cross-linking with secondary anti Fc antibody. Antibodies were used at concentrations of 7 nM
(black bars) and 0.7 nM (hatched bars). The vast majority of DRS binders (42 / 46) were able to induce DNA
fragmentation to different degrees after hyper-cross-linking.
Figure 14: Apoptosis induction in MDA-MB-231 cells measured via DNA
fragmentation ELISA assay by a selected series of novel DRS binders in the presence or absence of secondary, cross-linking anti Fc antibody to evaluate if the new DRS antibodies exhibit apoptosis activity without secondary cross-linking. The new DRS binders were compared to Drozitumab, an antibody known to be active to a certain degree without cross-linking.
Figure 15: Analysis of inhibition of cell proliferation (Cell TiterGlo Assay) of three different human tumor cells (DLD-1, NCI H460 and MDA-MB-231) upon treatment with different, cross-linked DRS antibodies at a concentration of 7 nM.
Figure 16: Evaluation of apoptosis induction measured by Caspase 8 activation in three human tumor cell lines (DLD-1, NCI H460 and MDA-MB-231) after treatment with cross-linked DRS
antibodies at a concentration of 7 nM.
Figure 17: Results of epitope binning experiments by Surface Plasmon Resonance (Biacore).
After binding of a first antibody to human DRS further binding of additional antibodies to be tested was analyzed. With the exception of clone 422, which might overlap with the Drozitumab epitope, none of the tested new DRS binders seem to bind to a region on DRS
that overlaps with Drozitumab epitope while among each other they might share at least overlapping epitopes.
Figure 18: TRAIL competition assay to determine ligand blocking vs. ligand non-blocking antibodies. Human TRAIL was immobilized on a CMS chip. Then a complex consisting of human DR5-Fc and DRS antibody was used as the analyte and binding of the complex to immobilized TRAIL was analyzed. While most of the new binders from phage display were TRAIL blocking molecules, a number of DRS antibodies from rabbit immunization were of the non-blocking kind.
-15-Figure 19: Effect of human TRAIL on inhibition of proliferation of DLD-1 target cells upon treatment with different DRS agonistic antibodies. DLD-1 target cells were incubated with cross-linked DRS antibodies in the absence (solid lines) or presence (stippled lines) of different concentrations of human TRAIL. Depending on the epitope recognized by the DRS
antibodies addition of TRAIL did (non-ligand blocking) or did not (ligand blocking) increase inhibition of cell growth. TRAIL alone and an antibody known to not block TRAIL were included as controls.
Figure 20: DNA fragmentation ELISA assay for detection of apoptosis. Apoptosis induction activity of different DRS-FAP bispecific antibodies (FAP clone 28H1) on MDA-MB-231 as the target cell line in the presence or absence of recombinant human FAP coated on a 96 well plate.
In this setting the coated FAP should mimic FAP expressed in the tumor stroma.
Bispecific antibodies were tested at two concentrations (7.0 and 0.7 nM). For the bispecific molecules containing the novel DRS binders a concentration dependent induction of apoptosis only was detected in the presence of FAP whereas the control molecule containing Drozitumab also showed some activity in the absence of FAP, confirming that the activity of the new DRS
antibodies is strictly dependent on cross-linking.
Figure 21: Bystander apoptosis induction activity of bispecific DRS-FAP
molecules (2+2 CrossFab format) in a co-culture assay using MDA-MB-231 as DRS expressing target cells and GM05389 (FAP fibroblasts) for cross-linking. Molecules containing different newly isolated DRS binders were compared to Drozitumab containing constructs at three concentrations (7.0, 0.7 and 0.07 nM). Under these conditions not all tested bispecific constructs exhibited the same activity compared to cross-linking via anti Fc mAb or via recombinant FAP.
This shows that not every antibody that can principally induce apoptosis after cross-linking also would do that under more natural conditions (in which the antigens-DRS and FAP-are expressed on different cell types).
Figure 22: Cell death detection ELISA (DNA fragmentation) with MDA-MB-231 target cells treated with new DRS binding Fabs fused to the C-terminus of human Fc, compared to Drozitumab, all after cross-linking with anti Fc antibody. All tested Fc fusion molecules are able to confer apoptosis induction on target cells in the same range as cross-linked Drozitumab, indicating that their binding and activity capacity is not blocked by positioning of the Fc to their N-terminus. A: DRS binders derived from phage display B: Humanized DRS binders derived from immunization.
Figure 23: Comparison of bispecific DRS-FAP antibodies (new DRS binders with C-terminal 28H1 CrossFab fusion) for apoptosis induction on MDA-MB-231 as the target cell line in a co-culture assay using GM05389 fibroblasts (DNA fragmentation ELISA assay for detection of apoptosis).
Figure 24: Bispecific DRS-FAP antibodies induce apoptosis on G401 cells in a bystander assay as determined by DNA fragmentation in a Cell Death Detection ELISA.
Figure 25: Schematic representation of the additional bispecific DRS-FAP
CrossFab formats that were evaluated with respect to productivity and quality. These four molecules differ in the position and valency of DRS and FAP binding moieties: two 2+1 molecules, one 3+1 and one
-16-1+1 construct were evaluated. One 2+1 molecule contains one FAP (28H1) CrossFab fused to the C-terminus of a DR5 (5E11) heavy chain (A) whereas a second 2+1 format consists of two DRS (5E11) Fabs fused to the N-terminus of an Fc with the 28H1 CrossMab counterpart (B).
The third molecule (C) contains three DRS targeting Fabs and one 28H1 CrossFab fused at the C-terminus of a heavy chain. In the 1+1 format (D) the DRS binder #174 is combined with the FAP binding moiety 4B9 in CrossFab arrangement. All constructs depend on hetero-dimerization using the knob-into-hole technology.
Figure 26: Apoptosis activity of bispecific DRS-FAP molecules (A: 5E11-28H1;
B: 18F11-28H1) in different formats as measured by Cell Death Detection ELISA for DNA
fragmentation in MDA-MB-231 cells co-cultured with GM05489 fibroblasts. The standard 2+2 format of Drozitumab-28H1 was compared to the 5E11-28H1 and 18F11-28H1 molecules in 2+2, 2+1 and 3+1 formats at three different concentrations.
Figure 27: Bystander apoptosis induction of the 5E11-28H1 molecule (2+2 format) compared to three additional bispecific formats: 2+1, novel 2+1 (head-to-tail fusion) and 3+1. MDA-MB-231 were used as target cells, GM05389 fibroblasts were co-cultured for FAP
dependent cross-linking and the bispecific antibodies were evaluated at concentrations from 700 -0.007 nM.
Figure 28: New bispecific 5E11-28H1 formats (2+2) for evaluation of productivity, side product profile and activity. The molecules differ in site and type of crossing.
A: C-terminal crossing of 28H1 (CH1CL) B: Crossing of entire N-terminal 5E11 Fab C: N-terminal VHVL crossing of 5E11 D: N-terminal CH1CL crossing of 5E11 E: Complete crossing of C-terminal 28H1 Fab F: Crossing of C-terminal 28H1 Fab (VHVL) Figure 29: FACS binding results with 5E11-28H1 CrossFabs in three different formats on MDA-MB-231 over a concentration range from 0.0037 to 60 nM. A PE conjugated goat-anti-human Fc (Fab)2 was used for detection.
Figure 30: DNA fragmentation ELISA assay for detection of apoptosis. Induction of apoptosis of 4 different CrossMab variants in co- (A) and mono-culture (B, C) settings as detected by DNA
fragmentation. All 4 different variants induce apoptosis in tumor cells in the co-culture setting in a comparable dose-dependent manner. In mono-culture settings no apoptosis is induced neither in MDA-MB231 tumor cell line nor in GM05389 fibroblast cell line, pointing out the specificity and FAP-dependency of apoptosis induction of all 4 variants.
Figure 31: In vivo efficacy in mouse xenograft tumor models in which DLD-1 (A;
nude mice) or MDA-MB-231 (B; SCID-beige mice) cells were co-injected with mu FAP expressing fibroblasts to ensure FAP expression in the tumor stroma. After the tumors have been engrafted treatment at 10 mg/kg was performed by i.v. injection of the bispecific Drozitumab-28H1 molecule (triangle) or Drozitumab alone (square) or the vehicle control diamond). Efficacy was determined by means of tumor growth inhibition (TGI) compared to the vehicle control.
Figure 32: Evaluation of bispecific DRS-FAP molecules produced for in vivo efficacy experiments for their apoptosis induction activity. The four different molecules were tested in a Cell Death Detection ELISA for DNA fragmentation of DLD-1 cells. 3T3 or recombinant 3T3
-17-cells expressing murine FAP wereused in the co-culture assay for cross-linking. Fold increase of apoptosis in comparison to untreated cells are shown.
Figure 33: Viability of DLD-1 cells after treatment with DR5-FAP bispecific antibodies after co-culture assay with 3T3 cells or 3T3 cells expressing murine FAP (Cell TiterGlo). Percentage of viability compared to an untreated control is given.
Figure 34: Mouse xenograft models in which different DRS-FAP bispecific molecules (10 mg/kg) in the 2+2 format were compared to a vehicle control. All constructs contained the 28H1 FAP CrossFab fused to different DRS binders: 5E11, 174 and 422. In addition one molecule (5E11-28H1) was included in which the Fc carried mutations to inhibit any FcyR
interaction (PGLALA). Efficacy was determined as tumor growth inhibition (TGI).
Figure 35: DNA fragmentation assay. Anti-DRS antibodies induce cell death upon receptor hyperclustering in a dose-dependent manner. The generated rabbit anti-DRS
antibodies were able to induce apoptosis of MDA-MB231 cells with different potencies but always in a dose-dependent fashion and only after Fc-mediated cross-linking of the DRS
molecules (upper panel).
In the absence of an anti-rabbit Fc-specific secondary antibody, no significant cell death was detected (lower panel).
Figure 36: Cell viability is diminished by anti-DRS antibodies upon receptor hyperclustering in a dose-dependent manner. The generated rabbit anti-DRS antibodies were able to decrease the viability of MDA-MB231 cells with varying potencies but always in a dose-dependent fashion and only after Fc-mediated cross-linking of the DRS molecules (upper panel).
In the absence of an anti-rabbit Fc-specific secondary antibody, the cell viability was not affected at any antibody concentration (lower panel).
Figure 37: Analysis of inhibition of cell proliferation (Cell TiterGlo Assay) of three different human tumor cells (DLD-1, NCI H460 and MDA-MB-231) upon treatment with different, cross-linked DRS antibodies at a concentration of 7 nM.
Figure 38: Evaluation of apoptosis induction measured by Caspase 8 activation in three human tumor cell lines (DLD-1, NCI H460 and MDA-MB-231) after treatment with cross-linked DRS
antibodies at a concentration of 7 nM.
Figure 39: Relative active concentration of stressed DRS antibody samples:
Exemplary response curve.
Figure 40: Relative active concentrations of original and stressed DRS
antibodies derived from immunization.
Figure 41: Relative active concentrations of original and stressed DRS
antibodies derived from phage display.
-18-Figure 42: Surface plasmon resonance (SPR, Biacore) analysis of simultaneous binding of different DRS-FAP bispecific antibodies to both recombinant targets. In a first reaction binding of the bispecific antibodies to recombinant human DRS-Fc was determined, followed by analysis of binding to recombinant human (upper panel)) or murine FAP (lower panel).
Figure 43: Induction of apoptosis in DLD-1 and H460 tumor cell lines by 2+2 bispecific constructs in co-culture assays as detected by DNA fragmentation. While a bispecific construct containing Drozitumab as DRS-binding component already induces apoptosis in the absence of FAP, all constructs containing new DRS binders derived by immunization only induce apoptosis in the presence of FAP. Constructs with newly developed DRS binders, such as 0011-28H1 and 0016-28H1, are able to induce apoptosis to a higher extent especially at low concentrations as compared to the Drozitumab containing bispecific construct.
Figure 44: Binding of anti-DRS-FAP bispecific 2+2 constructs to the DRS-expressing tumor cell line MDA-MB-231 as measured by flow cytometry analysis.
Figure 45: A Induction of apoptosis of humanized variants after crosslinking as detected by DNA fragmentation (Cell Death Detection ELISA): Humanized Variants of DRSTAA-(DRSTAA-0066 ¨ DRSTAA-0075, black lines) induce apoptosis upon crosslinking with secondary antibody in a dose-dependent manner. Several humanized variants, such as DRSTAA-0067, DRSTAA-0071, DRSTAA-0074 and DRSTAA-0075, are able to induce apoptosis in a similar manner concerning maximum of induction and dose-dependency as compared to the chimeric variant (DRSTAA-0052, grey lines). B Absence of induction of apoptosis of humanized variants without additional crosslinking as detected by DNA fragmentation (Cell Death Detection ELISA): Humanized Variants of DRSTAA-0011 (DRSTAA-0066 ¨ DRSTAA-0075) induce no apoptosis if not crosslinked by a secondary antibody.
Figure 46: Comparable induction of cell death in a co-culture system by bispecific anti-DRS-FAP antibodies in 1+1 and 2+2 formats.
Figure 47: Great specificity of bispecific anti-DRS-FAP antibodies at inducing cell death only in the presence of both tumor cells and fibroblasts.
Figure 48: Efficacy of bispecific anti-DR5-FAP (DR5 binder: VH SEQ ID NO.:7, VL SEQ ID
NO.: 8, FAP binder: VH SEQ ID NO.:15, VL SEQ ID NO.: 16) in DLD-1 CRC co-injection cell line based xenograft model Figure 49: Efficacy of bispecific anti-DRS-FAP (DRS binder: VH SEQ ID NO.:7, VL SEQ ID
NO.: 8, FAP binder: VH SEQ ID NO.:15, VL SEQ ID NO.: 16) in Co5896 CRC
fragment based patient derived xenograft model (PDX)
-19-DETAILED DESCRIPTION OF EMBODIMENTS OF THE INVENTION
I. DEFINITIONS
An "acceptor human framework" for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework "derived from" a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
"Affinity" refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., antibody and antigen).
The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
An "affinity matured" antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
The term "A bispecific antibody that specifically binds death receptor 5 (DR5) and Fibroblast Activation Protein (FAP)" refers to a bispecific antibody that is capable of binding DRS and FAP with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting cells expressing DRS and FAP). Specifically "A
bispecific antibody that specifically binds death receptor 5 (DRS) and Fibroblast Activation Protein (FAP)"
refers to a bispecific antibody targeting DRS on a tumor cell and FAP in the stroma surrounding said tumor. In one embodiment, the extent of binding of a bispecific antibody that specifically binds death receptor 5 (DRS) and Fibroblast Activation Protein (FAP) to an unrelated, non-FAP
-20-or non-DR5 protein is less than about 10% of the binding of the antibody to DRS or FAP as measured, e.g., by a Enzyme-linked immunosorbent assay (ELISA), surface plasmon resonance (SPR) based assays (e.g. Biacore) or flow cytometry (FACS). In certain embodiments, a bispecific antibody that specifically binds death receptor 5 (DRS) and Fibroblast Activation Protein (FAP) has a dissociation constant (Kd) of < 1 M, < 100 nM, < 10 nM, <
1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM (e.g. 10-8M or less, e.g. from 10-8M to 10-13M, e.g., from 10-9M to 10-13 M). In certain embodiments, a bispecific antibody that specifically binds death receptor 5 (DR5) and Fibroblast Activation Protein (FAP) binds to an epitope of DRS or FAP that is conserved among DRS or FAP from different species. Preferably said bispecific antibody binds to human and cynomolgous monkey DRS and to human, cynomolgous monkey and mouse FAP.
The terms "An antibody that specifically binds death receptor 5 (DRS)" refers to an antibody that is capable of binding DRS with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting cells expressing DRS. In one embodiment, the extent of binding of an antibody that specifically binds death receptor 5 (DRS) to an unrelated non-DRS protein is less than about 10% of the binding of the antibody to DRS as measured, e.g., by a radioimmunoassay (RIA) or flow cytometry (FACS). In certain embodiments, an antibody that specifically binds death receptor 5 (DRS) has a dissociation constant (Kd) of < 1 M, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or <
0.001 nM (e.g.
10-8M or less, e.g. from 10-8M to 10-13M, e.g., from 10-9M to 10-13 M). In certain embodiments, an antibody that specifically binds death receptor 5 (DRS) binds to an epitope of DRS that is conserved among DRS from different species. Preferably said antibody binds to human and cynomolgous monkey DRS. The term "An antibody that specifically binds death receptor 5 (DRS)" also encompasses bispecific antibodies that are capable of binding DRS
and a second antigen.
The term "antibody" herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2;
diabodies, cross-Fab fragments; linear antibodies; single-chain antibody molecules (e.g. scFv);
and multispecific antibodies formed from antibody fragments. scFv antibodies are, e.g. described
-21-in Houston, J.S., Methods in Enzymol. 203 (1991) 46-96). In addition, antibody fragments comprise single chain polypeptides having the characteristics of a VH domain, namely being able to assemble together with a VL domain, or of a VL domain, namely being able to assemble together with a VH domain to a functional antigen binding site and thereby providing the antigen binding property of full length antibodies.
As used herein, "Fab fragment" refers to an antibody fragment comprising a light chain fragment comprising a VL domain and a constant domain of a light chain (CL), and a VH
domain and a first constant domain (CH1) of a heavy chain. In one embodiment the bispecific antibodies of the invention comprise at least one Fab fragment, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
Due to the exchange of either the variable regions or the constant regions, said Fab fragment is also referred to as "cross-Fab fragment" or "xFab fragment" or "crossover Fab fragment". Two different chain compositions of a crossover Fab molecule are possible and comprised in the bispecific antibodies of the invention: On the one hand, the variable regions of the Fab heavy and light chain are exchanged, i.e. the crossover Fab molecule comprises a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1), and a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL). This crossover Fab molecule is also referred to as CrossFab (VH). On the other hand, when the constant regions of the Fab heavy and light chain are exchanged, the crossover Fab molecule comprises a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL), and a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1). This crossover Fab molecule is also referred to as Cros s Fab (cLat p=
A "single chain Fab fragment" or "scFab" is a polypeptide consisting of an antibody heavy chain variable domain (VH), an antibody constant domain 1 (CH1), an antibody light chain variable domain (VL), an antibody light chain constant domain (CL) and a linker, wherein said antibody domains and said linker have one of the following orders in N-terminal to C-terminal direction:
a) VH-CH1-linker-VL-CL, b) VL-CL-linker-VH-CH1, c) VH-CL-linker-VL-CH1 or d) VL-CH1-linker-VH-CL; and wherein said linker is a polypeptide of at least 30 amino acids, preferably between 32 and 50 amino acids. Said single chain Fab fragments a) VH-CH1-linker-VL-CL, b) VL-CL-linker-VH-CH1, c) VH-CL-linker-VL-CH1 and d) VL-CH1-linker-VH-CL, are stabilized via the natural disulfide bond between the CL domain and the CH1 domain. In addition, these single chain Fab molecules might be further stabilized by generation of interchain disulfide bonds via insertion of cysteine residues (e.g. position 44 in the variable heavy chain and positionn 100 in the variable light chain according to Kabat numbering). The term "N-terminus
-22-denotes the last amino acid of the N-terminus. The term "C-terminus denotes the last amino acid of the C-terminus.
By "fused" or "connected" is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.
The term "linker" as used herein refers to a peptide linker and is preferably a peptide with an amino acid sequence with a length of at least 5 amino acids, preferably with a length of 5 to 100, more preferably of 10 to 50 amino acids. In one embodiment said peptide linker is (GxS)n or (GxS)nGm with G = glycine, S = serine, and (x = 3, n= 3, 4, 5 or 6, and m=
0, 1, 2 or 3) or (x = 4,n= 2, 3, 4 or 5 and m= 0, 1, 2 or 3), preferably x = 4 and n= 2 or 3, more preferably with x =
4, n= 2. In one embodiment said peptide linker is (G4S)2.
The term "immunoglobulin molecule" refers to a protein having the structure of a naturally occurring antibody. For example, immunoglobulins of the IgG class are heterotetrameric glycoproteins of about 150,000 daltons, composed of two light chains and two heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3), also called a heavy chain constant region.
Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain, also called a light chain constant region. The heavy chain of an immunoglobulin may be assigned to one of five types, called a (IgA), 6 (IgD), e (IgE), 7 (IgG), or (IgM), some of which may be further divided into subtypes, e.g. 71 (IgGO, 72 (IgG2), 73 (IgG3), 74 (Igat), al (IgAO and L2 (IgA2). The light chain of an immunoglobulin may be assigned to one of two types, called kappa (x) and lambda (4 based on the amino acid sequence of its constant domain. An immunoglobulin essentially consists of two Fab molecules and an Fc domain, linked via the immunoglobulin hinge region.
An "antibody that binds to the same epitope" as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more. An exemplary competition assay is provided herein.
The term "antigen binding domain" refers to the part of an antigen binding molecule that comprises the area which specifically binds to and is complementary to part or all of an antigen.
Where an antigen is large, an antigen binding molecule may only bind to a particular part of the
-23-antigen, which part is termed an epitope. An antigen binding domain may be provided by, for example, one or more antibody variable domains (also called antibody variable regions).
Preferably, an antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).
The term "chimeric" antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising a rabbit variable region and a human constant region are preferred. Other preferred forms of "chimeric antibodies"
encompassed by the present invention are those in which the constant region has been modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to Clq binding and/or Fc receptor (FcR) binding. Such chimeric antibodies are also referred to as "class-switched antibodies". Chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding immunoglobulin variable regions and DNA
segments encoding immunoglobulin constant regions. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art. See e.g. Morrison, S.L., et al., Proc. Natl. Acad. Sci. USA
81 (1984) 6851-6855; US Patent Nos. 5,202,238 and 5,204,244.
The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At2I I , 1131, 1125, y90, Re186 , Re188 , Sm153 , Bi212 , P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.
"Effector functions" refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include:
Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding;
antibody-dependent cell-mediated cytotoxicity (ADCC); antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells; down regulation of cell surface receptors (e.g. B cell receptor); and B
cell activation.
-24-As used herein, the terms "engineer, engineered, engineering", are considered to include any manipulation of the peptide backbone or the post-translational modifications of a naturally occurring or recombinant polypeptide or fragment thereof. Engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches.
The term "amino acid mutation" as used herein is meant to encompass amino acid substitutions, deletions, insertions, and modifications. Any combination of substitution, deletion, insertion, and modification can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., reduced binding to an Fc receptor, or increased association with another peptide. Amino acid sequence deletions and insertions include amino- and/or carboxy-terminal deletions and insertions of amino acids.
Particular amino acid mutations are amino acid substitutions. For the purpose of altering e.g. the binding characteristics of an Fc region, non-conservative amino acid substitutions, i.e. replacing one amino acid with another amino acid having different structural and/or chemical properties, are particularly preferred. Amino acid substitutions include replacement by non-naturally occurring amino acids or by naturally occurring amino acid derivatives of the twenty standard amino acids (e.g. 4-hydroxyproline, 3-methylhistidine, ornithine, homoserine, 5-hydroxylysine). Amino acid mutations can be generated using genetic or chemical methods well known in the art. Genetic methods may include site-directed mutagenesis, PCR, gene synthesis and the like. It is contemplated that methods of altering the side chain group of an amino acid by methods other than genetic engineering, such as chemical modification, may also be useful.
Various designations may be used herein to indicate the same amino acid mutation. For example, a substitution from proline at position 329 of the Fc domain to glycine can be indicated as 329G, G329, G329, P329G, or Pro329Gly.
An "effective amount" of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
The term "Fc domain" or "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. Although the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to extend from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present.
-25-Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991. A "subunit" of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association.
For example, a subunit of an IgG Fc domain comprises an IgG CH2 and an IgG CH3 constant domain.
A "modification promoting the association of the first and the second subunit of the Fc domain" is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer. A
modification promoting association as used herein particularly includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the modifications are complementary to each other so as to promote association of the two Fc domain subunits. For example, a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their association sterically or electrostatically favorable, respectively. Thus, (hetero)dimerization occurs between a polypeptide comprising the first Fc domain subunit and a polypeptide comprising the second Fc domain subunit, which might be non-identical in the sense that further components fused to each of the subunits (e.g. antigen binding moieties) are not the same. In some embodiments the modification promoting association comprises an amino acid mutation in the Fc domain, specifically an amino acid substitution. In a particular embodiment, the modification promoting association comprises a separate amino acid mutation, specifically an amino acid substitution, in each of the two subunits of the Fc domain.
"Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR
domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1 -H1(L1)-FR2-H2(L2)-FR3-H3 (L3)-FR4.
The terms "full length antibody," "intact antibody," and "whole antibody" are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
-26-The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells,"
which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
A "human antibody" is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues. As also mentioned for chimeric and humanized antibodies according to the invention the term "human antibody" as used herein also comprises such antibodies which are modified in the constant region to generate the properties according to the invention, especially in regard to C 1 q binding and/or FcR binding, e.g. by "class switching" i.e.
change or mutation of Fc parts (e.g. from IgG1 to IgG4 and/or IgGl/IgG4 mutation.) The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NSO or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell. Such recombinant human antibodies have variable and constant regions in a rearranged form. The recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation. Thus, the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germ line VH and VL sequences, may not naturally exist within the human antibody germ line repertoire in vivo.
A "human consensus framework" is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH
framework sequences. Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences. Generally, the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH Publication 91-3242, Bethesda MD (1991), vols. 1-3. In one embodiment, for the VL, the subgroup is
-27-subgroup kappa I as in Kabat et al., supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al., supra.
A "humanized" antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs. In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization. Other forms of "humanized antibodies" encompassed by the present invention are those in which the constant region has been additionally modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to Clq binding and/or Fc receptor (FcR) binding.
The term "hypervariable region" or "HVR," as used herein refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops"). Generally, native four-chain antibodies comprise six HVRs; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the "complementarity determining regions" (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. Exemplary hypervariable loops occur at amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3). (Chothia and Lesk, J. Mol. Biol.
196:901-917 (1987).) Exemplary CDRs (CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3) occur at amino acid residues 24-34 of L 1, 50-56 of L2, 89-97 of L3, 31-35B of H1, 50-65 of H2, and 95-102 of H3. (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD
(1991).) Hypervariable regions (HVRs) are also referred to as complementarity determining regions (CDRs), and these terms are used herein interchangeably in reference to portions of the variable region that form the antigen binding regions. This particular region has been described by Kabat et al., U.S. Dept. of Health and Human Services, "Sequences of Proteins of Immunological Interest" (1983) and by Chothia et al., J. Mol. Biol. 196:901-917 (1987), where the definitions include overlapping or subsets of amino acid residues when compared against each other.
Nevertheless, application of either definition to refer to a CDR of an antibody or variants thereof
-28-is intended to be within the scope of the term as defined and used herein. The appropriate amino acid residues which encompass the CDRs as defined by each of the above cited references are set forth below in Table A as a comparison. The exact residue numbers which encompass a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can routinely determine which residues comprise a particular CDR given the variable region amino acid sequence of the antibody.
TABLE A. CDR Definitionsl CDR Kabat Chothia AbM2 1Numbering of all CDR definitions in Table A is according to the numbering conventions set forth by Kabat et al. (see below).
2 "AbM" with a lowercase "b" as used in Table A refers to the CDRs as defined by Oxford Molecular's "AbM" antibody modeling software.
Kabat et al. also defined a numbering system for variable region sequences that is applicable to any antibody. One of ordinary skill in the art can unambiguously assign this system of "Kabat numbering" to any variable region sequence, without reliance on any experimental data beyond the sequence itself. As used herein, "Kabat numbering" refers to the numbering system set forth by Kabat et al., U.S. Dept. of Health and Human Services, "Sequence of Proteins of Immunological Interest" (1983). Unless otherwise specified, references to the numbering of specific amino acid residue positions in an antibody variable region are according to the Kabat numbering system.
With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. CDRs also comprise "specificity determining residues," or "SDRs," which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a-CDRs. Exemplary a-CDRs (a-CDR-L1, a-CDR-L2, a-CDR-L3, a-CDR-H1, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of Ll, 50-55 of L2, 89-96 of L3, 31-35B of H1, 50-58 of H2, and 95-102 of H3. (See Almagro and Fransson, Front.
Biosci. 13:1619-1633 (2008).) Unless otherwise indicated, HVR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Kabat et al., supra.
An "immunoconjugate" is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
-29-An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
In certain embodiments, the individual or subject is a human.
An "isolated" antibody is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95%
or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman et al., J. Chromatogr. B
848:79-87 (2007).
An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
"Isolated nucleic acid encoding a bispecific antibody that specifically binds DR5 and FAP antibody" refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. Thus, the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and
-30-methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
A "naked antibody" refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.
"Native antibodies" refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (x) and lambda (4 based on the amino acid sequence of its constant domain.
The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
"No substantial cross-reactivity" means that a molecule (e.g., an antibody) does not recognize or specifically bind an antigen different from the actual target antigen of the molecule (e.g. an antigen closely related to the target antigen), particularly when compared to that target antigen. For example, an antibody may bind less than about 10% to less than about 5% to an antigen different from the actual target antigen, or may bind said antigen different from the actual target antigen at an amount consisting of less than about 10%, 9%, 8%
7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.2%, or 0.1%, preferably less than about 2%, 1%, or 0.5%, and most preferably less than about 0.2% or 0.1% antigen different from the actual target antigen.
"Percent (%) amino acid sequence identity" with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment
-31-for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
In situations where ALIGN-2 is employed for amino acid sequence comparisons, the %
amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A
that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction X/Y
where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y
is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program.
The term "pharmaceutical formulation" refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
-32-A "pharmaceutically acceptable carrier" refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
The term "death receptor 5 (DR5)", as used herein, refers to any native DRS
from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated. The term encompasses "full-length,"
unprocessed DRS as well as any form of DRS that results from processing in the cell. The term also encompasses naturally occurring variants of DRS, e.g., splice variants or allelic variants. The amino acid sequence of an exemplary human DRS is shown in SEQ ID NO.:155.
The term "Fibroblast activation protein (FAP)", as used herein, refers to any native FAP
from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated. The term encompasses "full-length," unprocessed FAP as well as any form of FAP that results from processing in the cell. The term also encompasses naturally occurring variants of FAP, e.g., splice variants or allelic variants.
Preferably, an anti-FAP antibody of the invention binds to the extracellular domain of FAP. The amino acid sequence of exemplary human, mouse and cynomolgus monkey FAP
ectodomains (with a C-terminal poly-lysine and 6x His-tag) are shown in SEQ ID NO.:156, SEQ ID NO.:157, and SEQ ID NO.:158 respectively.
As used herein, "treatment" (and grammatical variations thereof such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
In some embodiments, antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
The term cancer as used herein refers to proliferative diseases, such as lymphomas, lymphocytic leukemias, lung cancer, non small cell lung (NSCL) cancer, bronchioloalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the
-33-vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, mesothelioma, hepatocellular cancer, biliary cancer, neoplasms of the central nervous system (CNS), spinal axis tumors, brain stem glioma, glioblastoma multiforme, astrocytomas, schwanomas, ependymonas, medulloblastomas, meningiomas, squamous cell carcinomas, pituitary adenoma and Ewings sarcoma, including refractory versions of any of the above cancers, or a combination of one or more of the above cancers.
The term "variable region" or "variable domain" refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). (See, e.g., Kindt et al. Kuby Immunology, 6th ed., W.H.
Freeman and Co., page 91 (2007).) A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J.
Immunol.
150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
The term "antigen-binding site of an antibody" when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding. The antigen-binding portion of an antibody comprises amino acid residues from the "complementary determining regions" or "CDRs". "Framework" or "FR" regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chain variable domains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. Especially, CDR3 of the heavy chain is the region which contributes most to antigen binding and defines the antibody's properties. CDR and FR regions are determined according to the standard definition of Kabat et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991) and/or those residues from a "hypervariable loop".
Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Natural antibodies, for example, are monospecific.
"Bispecific antibodies"
according to the invention are antibodies which have two different antigen-binding specificities.
-34-Antibodies of the present invention are specific for two different antigens, i.e. DR5 as first antigen and FAP as second antigen.
The term "monospecific" antibody as used herein denotes an antibody that has one or more binding sites each of which bind to the same epitope of the same antigen.
The term "bispecific" antibody as used herein denotes an antibody that has at least two binding sites each of which bind to different epitopes of the same antigen or a different antigen.
The antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. Provided herein is a bispecific antibody, with binding specificities for FAP and DRS. In certain embodiments, bispecific antibodies may bind to two different epitopes of DRS.
Bispecific antibodies may also be used to localize cytotoxic agents to cells which express DRS.
Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)), WO
93/08829, and Traunecker et al., EMBO J. 10: 3655 (1991)), and "knob-in-hole" engineering (see, e.g., U.S.
Patent No. 5,731,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO
2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No.
4,676,980, and Brennan et al., Science, 229: 81 (1985)); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny et al., J. Immunol., 148(5):1547-1553 (1992)); using "diabody"
technology for making bispecific antibody fragments (see, e.g., Hollinger et al., Proc. Natl. Acad.
Sci. USA, 90:6444-6448 (1993)); and using single-chain Fv (sFv) dimers (see,e.g. Gruber et al., J. Immunol., 152:5368 (1994)); and preparing trispecific antibodies as described, e.g., in Tutt et al. J.
Immunol. 147: 60 (1991).
Engineered antibodies with three or more functional antigen binding sites, including "Octopus antibodies," are also included herein (see, e.g. US 2006/0025576A1).
The antibody or fragment herein also includes a "Dual Acting FAb" or "DAF"
comprising at least one antigen binding site that binds to FAP or DRS as well as another, different antigen (see, US 2008/0069820, for example).
The term "valent" as used within the current application denotes the presence of a specified number of binding sites in an antibody molecule. As such, the terms "bivalent", "tetravalent", and "hexavalent" denote the presence of two binding sites, four binding sites, and
-35-six binding sites, respectively, in an antibody molecule. The bispecific antibodies according to the invention are at least "bivalent" and may be "trivalent" or "multivalent"
(e.g."tetravalent" or "hexavalent").
Antibodies of the present invention have two or more binding sites and are bispecific.
That is, the antibodies may be bispecific even in cases where there are more than two binding sites (i.e. that the antibody is trivalent or multivalent). Bispecific antibodies of the invention include, for example, multivalent single chain antibodies, diabodies and triabodies, as well as antibodies having the constant domain structure of full length antibodies to which further antigen-binding sites (e.g., single chain Fv, a VH domain and/or a VL domain, Fab, or (Fab)2) are linked via one or more peptide-linkers. The antibodies can be full length from a single species, or be chimerized or humanized.
The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors."
The term "amino acid" as used within this application denotes the group of naturally occurring carboxy a-amino acids comprising alanine (three letter code: ala, one letter code: A), arginine (arg, R), asparagine (asn, N), aspartic acid (asp, D), cysteine (cys, C), glutamine (gln, Q), glutamic acid (glu, E), glycine (gly, G), histidine (his, H), isoleucine (ile, I), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y), and valine (val, V).
As used herein, the expressions "cell", "cell line", and "cell culture" are used interchangeably and all such designations include progeny. Thus, the words "transfectants" and "transfected cells" include the primary subject cell and cultures derived there from without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included.
"Affinity" refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity
-36-which reflects a 1:1 interaction between members of a binding pair (e.g., antibody and antigen).
The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
As used herein, the term "binding" or "specifically binding" refers to the binding of the antibody to an epitope of the antigen in an in-vitro assay, preferably in a surface plasmon resonance assay (SPR, BIAcore, GE-Healthcare Uppsala, Sweden). The affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kD (dissociation constant), and KD (kD/ka). Binding or specifically binding means a binding affinity (KD) of 10-8 mo1/1 or less, preferably 10-9 M
to 10-13 mo1/1.
Binding of the antibody to the death receptor can be investigated by a BIAcore assay (GE-Healthcare Uppsala, Sweden). The affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kD (dissociation constant), and KD (kD/ka) The term "epitope" includes any polypeptide determinant capable of specific binding to an antibody. In certain embodiments, epitope determinant include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and or specific charge characteristics. An epitope is a region of an antigen that is bound by an antibody.
As used herein, the terms "engineer, engineered, engineering," particularly with the prefix "glyco-," as well as the term "glycosylation engineering" are considered to include any manipulation of the glycosylation pattern of a naturally occurring or recombinant polypeptide or fragment thereof. Glycosylation engineering includes metabolic engineering of the glycosylation machinery of a cell, including genetic manipulations of the oligosaccharide synthesis pathways to achieve altered glycosylation of glycoproteins expressed in cells.
Furthermore, glycosylation engineering includes the effects of mutations and cell environment on glycosylation. In one embodiment, the glycosylation engineering is an alteration in glycosyltransferase activity. In a particular embodiment, the engineering results in altered glucosaminyltransferase activity and/or fucosyltransferase activity.
-37-II. COMPOSITIONS AND METHODS
In one aspect, the invention is based on bispecific antibodies comprising a first antigen binding site specific for TRAIL death receptor 5 (DR5) and a second antigen binding site specific for Fibroblast Activation Protein (FAP). In another embodiment novel antibodies targeting DRS are provided. Antibodies of the invention are useful, e.g., for the treatment or diagnosis of cancer.
A. Exemplary bispecific antibodies that bind to DR5 and FAP
In one aspect, the invention provides isolated bispecific antibodies that bind to DRS and FAP. FAP binding moieties have been described in WO 2012/020006, which is included by reference in its entirety. FAP binding moieties of particular interest to be used in the DR5-FAP
bispecific antibodies are outlined in the embodiments below.
In certain embodiments, a bispecific antibody that binds to DRS and FAP
specifically crosslinks the death receptors and apoptosis of the target cell is induced.
The advantage of these bispecific death receptor agonistic antibodies over conventional death receptor targeting antibodies is the specificity of induction of apoptosis only at the site where FAP is expressed. As outlined above the inventors of the present invention developed novel DRS
binding moieties with superior properties compared to known DRS binders that can be incorporated into novel and advantageous DR5-FAP bispecific antibodies.
In one aspect, the invention provides a bispecific antibody that binds to death receptor 5 (DRS) and Fibroblast Activation Protein (FAP) comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 consisting of SEQ ID NO.:1, SEQ ID NO.:17 and SEQ ID
NO.:75;
(b) a heavy chain CDR2 of SEQ ID NO.:2, SEQ ID NO.:18, SEQ ID NO.:25 and SEQ
ID
NO.:83;
(c) a heavy chain CDR3 of SEQ ID NO. :3, SEQ ID NO.:19, SEQ ID NO. :84, SEQ ID
NO. :96, SEQ ID NO. :98, SEQ ID NO.:104 and SEQ ID NO.:108;
(d) a light chain CDR1 of SEQ ID NO. :4, SEQ ID NO. :20, SEQ ID NO. :27 and SEQ ID
NO.:86;
(e) a light chain CDR2 of SEQ ID NO. :5, SEQ ID NO. :21 and SEQ ID NO. :28;
and (f) a light chain CDR3 of SEQ ID NO. :6, SEQ ID NO. :22, SEQ ID NO. :87, SEQ
ID NO. :99, SEQ ID NO.:105, SEQ ID NO.:109 and SEQ ID NO. :97;
-38-and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO.:9 and SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:10 and SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO.:11 and SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO.:12 and SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO.:13 and SEQ ID NO.:37;
(f) a light chain CDR3 of SEQ ID NO.:14 and SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO.:6 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
-39-(f) a light chain CDR3 of SEQ ID NO.:6 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO.:21; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
-40-(e) a light chain CDR2 of SEQ ID NO.:21; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO.:21; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
-41-(e) a light chain CDR2 of SEQ ID NO.:21; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :27;
(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :27;
-42-(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
-43-(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
-44-(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO. :75;
(b) a heavy chain CDR2 of SEQ ID NO. :83;
(c) a heavy chain CDR3 of SEQ ID NO. :84;
(d) a light chain CDR1 of SEQ ID NO. :86;
(e) a light chain CDR2 of SEQ ID NO. :28;
(f) a light chain CDR3 of SEQ ID NO.:87 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO. :75;
(b) a heavy chain CDR2 of SEQ ID NO. :83;
(c) a heavy chain CDR3 of SEQ ID NO.:84;
(d) a light chain CDR1 of SEQ ID NO. :86;
45 (e) a light chain CDR2 of SEQ ID NO. :28;
(f) a light chain CDR3 of SEQ ID NO.:87 and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO. :34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DR5, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO. :96;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO. :99;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:96;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
-46-(f) a light chain CDR3 of SEQ ID NO. :99;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:104;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO:105;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:104;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
-47-(f) a light chain CDR3 of SEQ ID NO:105;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DR5, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:108;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO:109;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:108;
-48-(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO:109;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO.:33;
(b) a heavy chain CDR2 of SEQ ID NO.:34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DR5, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO. :98;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO. :97;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DRS
and FAP
comprising at least one antigen binding site specific for DRS, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
-49-(c) a heavy chain CDR3 of SEQ ID NO. :98;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO. :97;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DR5, comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :27;
(e) a light chain CDR2 of SEQ ID NO. :28;
(f) a light chain CDR3 of SEQ ID NO. :22;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
In one aspect, the invention provides a bispecific antibody that binds to DR5 and FAP
comprising at least one antigen binding site specific for DRS, comprising
-50-(a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :27;
(e) a light chain CDR2 of SEQ ID NO. :28;
(f) a light chain CDR3 of SEQ ID NO. :22;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 of SEQ ID NO. :33;
(b) a heavy chain CDR2 of SEQ ID NO.:34;
(c) a heavy chain CDR3 of SEQ ID NO. :35;
(d) a light chain CDR1 of SEQ ID NO. :36;
(e) a light chain CDR2 of SEQ ID NO. :37;
(f) a light chain CDR3 of SEQ ID NO. :38.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5 comprising a variable heavy chain and a variable light chain comprising an amino acid sequence selected from the group of: SEQ ID NO. :7 and SEQ ID NO.
:8; SEQ ID
NO.:23 and SEQ ID NO.:24; SEQ ID NO. :26 and SEQ ID NO. :24; SEQ ID NO.:23 and SEQ ID
NO.:29; SEQ ID NO.:23 and SEQ ID NO.:30; SEQ ID NO.:26 and SEQ ID NO.:31; SEQ
ID
NO. :26 and SEQ ID NO.:32; SEQ ID NO. :26 and SEQ ID NO. :30; SEQ ID NO.:23 and SEQ ID
NO.:31; SEQ ID NO.:82 and SEQ ID NO.:85; SEQ ID NO.:100 and SEQ ID NO.:101;
SEQ ID
NO.:102 and SEQ ID NO.:103; SEQ ID NO.:106 and SEQ ID NO.:107; SEQ ID NO. :94 and SEQ ID NO. :95;
and at least one antigen binding site specific for FAP comprisings a variable heavy chain comprising an amino acid sequence selected from the group of: SEQ ID NO.:15 and SEQ ID
NO.:39 ; and a light chain variable region comprising an amino acid sequence selected from the group of SEQ ID NO.:16 and SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :7 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :8; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region
-51-comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :7 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :8; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :24; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :24; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :24; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
-52-ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :24; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :29; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :29; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:30; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:30; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
-53-In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:31; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:31; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:32; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:32; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:30; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region
-54-comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:30; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:31; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:31; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :82 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :85; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
-55-ID NO. :82 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :85; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:100 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:101;
and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:100 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:101;
and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:102 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:103;
and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:102 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:103;
and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
-56-In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DR5, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:106 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:107;
and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO.:106 and a variable light chain comprising an amino acid sequence of SEQ
ID NO.:107;
and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :94 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :95; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
In one embodiment, the bispecific antibody comprises at least one antigen binding site specific for DRS, comprising a variable heavy chain comprising an amino acid sequence of SEQ
ID NO. :94 and a variable light chain comprising an amino acid sequence of SEQ
ID NO. :95; and at least one antigen binding site specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:39 and a light chain variable region comprising an amino acid sequence of SEQ ID NO. :40.
In one embodiment, the bispecific antibody of the invention comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID NO.:151.
In one embodiment, the bispecific antibody of the invention comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID NO.:152.
-57-In another embodiment the bispecific antibody of the invention comprises a light chain constant region comprising the amino acid sequence of SEQ ID NO.:153.
In one embodiment, the bispecific antibody of the invention comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID NO.:151, wherein the C-terminal Lysine has been removed.
In one embodiment, the bispecific antibody of the invention comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID NO.:152, wherein the C-terminal Lysine has been removed.
In one embodiment the bispecific antibody of the invention comprises a first antibody comprising at least one antigen binding site specific for DR5, said first antibody comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8, and a heavy chain constant region comprising the amino acid sequence selected from of SEQ
ID NO.:151 or SEQ ID NO.:152, and a light chain constant region comprising the amino acid sequence of SEQ
ID NO.:153, and a second antibody specific for FAP comprising one or more amino acid sequences as defined in any of the embodiments above. In one embodiment the C-terminal Lysine of the amino acid sequence of said heavy chain constant region has been removed.
In one embodiment the bispecific antibody of the invention comprises a first antibody comprising at least one antigen binding site specific for DR5, said first antibody comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8, and a heavy chain constant region comprising the amino acid sequence selected from of SEQ
ID NO.:151 or SEQ ID NO.:152, and a light chain constant region comprising the amino acid sequence of SEQ
ID NO.:153, and a second antibody specific for FAP comprising a variable heavy chain of SEQ
ID NO.:15 and a variable light chain of SEQ ID NO.:16.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:131, SEQ
ID NO.:132 and SEQ ID NO.:124.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:133, SEQ
ID NO.:132 and SEQ ID NO.:124.
In one preferred embodiment a bispecific antibody is provided comprising SEQ
ID
NO.:134, SEQ ID NO.:132 and SEQ ID NO.:124.
-58-In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:262, SEQ
ID NO.:263 and SEQ ID NO.:132.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:135, SEQ
ID NO.:136 and SEQ ID NO.:137.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:138, SEQ
ID NO.:139 and SEQ ID NO.:137.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:274, SEQ
ID NO.:275, and SEQ ID NO.:137.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:276, SEQ
ID NO.:277, and SEQ ID NO.:132.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:278, SEQ
ID NO.:279 and SEQ ID NO.:132.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:142, SEQ
ID NO.:143, SEQ ID NO.:124 and SEQ ID NO.:132.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:145, SEQ
ID NO.:146, SEQ ID NO.:124 and SEQ ID NO.:132.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:144, SEQ
ID NO.:143, SEQ ID NO.:124 and SEQ ID NO.:132.
In one embodiment a bispecific antibody is provided comprising SEQ ID NO.:159, SEQ
ID NO.:160, SEQ ID NO.:161 and SEQ ID NO.:162.
In another aspect, a bispecific antibody that binds to DR5 and FAP comprises at least one antigen binding site specific for DR5 comprising a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%
sequence identity to the amino acid sequence of SEQ ID NO. :7, and at least one antigen binding site specific for FAP comprising a variable heavy chain of SEQ ID NO.:15 and a variable light chain of SEQ ID NO.:16.
In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions),
-59-insertions, or deletions relative to the reference sequence, but a bispecific antibody that binds to DR5 and FAP comprising that sequence retains the ability to bind to FAP and DRS. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO. :7. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the bispecific antibody that binds to DRS and FAP comprises the VH sequence in SEQ ID NO. :7, including post-translational modifications of that sequence.
In another aspect, a bispecific antibody that binds to DRS and FAP comprises at least one antigen binding site specific for DRS comprising a light chain variable domain (VL) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%
sequence identity to the amino acid sequence of SEQ ID NO.:8, and at least one antigen binding site specific for FAP comprising a variable heavy chain of SEQ ID NO.:15 and a variable light chain of SEQ ID NO.:16.
In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but a bispecific antibody that binds to DRS and FAP comprising that sequence retains the ability to bind to DRS and FAP. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO.:8. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the bispecific antibody that binds to DRS
and FAP comprises the VL sequence in SEQ ID NO:8, including post-translational modifications of that sequence.
In another aspect, a bispecific antibody that binds to DRS and FAP is provided, comprising at least one antigen binding site specific for DRS comprising a variable light chain of SEQ ID NO. :8 and a variable heavy chain of SEQ ID NO. :7; and at least one antigen binding site specific for FAP, comprising a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO.:15. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but a bispecific antibody that binds to DRS and FAP comprising that sequence retains the ability to bind to FAP and DRS. In certain embodiments, a total of 1 to 10 amino acids have been
-60-substituted, inserted and/or deleted in SEQ ID NO.:15. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
Optionally, the bispecific antibody that binds to DR5 and FAP comprises the VH sequence in SEQ
ID NO.:15, including post-translational modifications of that sequence.
In another aspect, a bispecific antibody that binds to DRS and FAP is provided, comprising at least one antigen binding site specific for DRS, comprising a variable light chain of SEQ ID NO.:8 and a variable heavy chain of SEQ ID NO. :7, and at least one antigen binding site specific for FAP, comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO.:16. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but a bispecific antibody that binds to DRS and FAP comprising that sequence retains the ability to bind to DRS and FAP. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO.:16. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
Optionally, the bispecific antibody that binds to DRS and FAP comprises the VL sequence in SEQ
ID NO:16, including post-translational modifications of that sequence.
In another aspect, a a bispecific antibody that binds to DRS and FAP is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above. In one embodiment, the antibody comprises the VH and VL
sequences in SEQ ID NO:7 and SEQ ID NO:8, and SEQ ID NO:15 and SEQ ID NO:16, respectively, including post-translational modifications of those sequences.
In a further aspect of the invention, a bispecific antibody that binds to DRS
and FAP
according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody. In one embodiment said bispecific antibody that binds to DRS
and FAP according to any of the above embodiments is a human antibody.
-61-B. Exemplary antibodies that bind to DR5 In one aspect, the invention provides isolated antibodies and antibody fragments that bind to DR5. These death receptor agonistic antibodies have superior properties compared to known DRS binders that can be incorporated into novel and advantageous bispecific antibodies targeting DRS and a second antigen.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DRS), comprising (a) a heavy chain CDR1 consisting of SEQ ID NO.:1, SEQ ID NO.:17 and SEQ ID
NO.:75;
(b) a heavy chain CDR2 of SEQ ID NO.:2, SEQ ID NO.:18, SEQ ID NO.:25 and SEQ
ID
NO.:83;
(c) a heavy chain CDR3 of SEQ ID NO. :3, SEQ ID NO.:19, SEQ ID NO. :84, SEQ ID
NO. :96, SEQ ID NO. :98, SEQ ID NO.:104 and SEQ ID NO.:108;
(d) a light chain CDR1 of SEQ ID NO. :4, SEQ ID NO. :20, SEQ ID NO. :27 and SEQ ID
NO.:86;
(e) a light chain CDR2 of SEQ ID NO.:5, SEQ ID NO.:21 and SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :6, SEQ ID NO. :22, SEQ ID NO. :87, SEQ
ID NO. :99, SEQ ID NO.:105, SEQ ID NO.:109 and SEQ ID NO. :97.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DRS), comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO.:6.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DRS), comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
-62-(e) a light chain CDR2 of SEQ ID NO.:21; and (f) a light chain CDR3 of SEQ ID NO. :22.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO.:21; and (f) a light chain CDR3 of SEQ ID NO. :22.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :27;
(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO.:18;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising
-63-(a) a heavy chain CDR1 of SEQ ID NO.:17;
(b) a heavy chain CDR2 of SEQ ID NO. :25;
(c) a heavy chain CDR3 of SEQ ID NO.:19;
(d) a light chain CDR1 of SEQ ID NO. :20;
(e) a light chain CDR2 of SEQ ID NO. :28; and (f) a light chain CDR3 of SEQ ID NO. :22.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO. :75;
(b) a heavy chain CDR2 of SEQ ID NO. :83;
(c) a heavy chain CDR3 of SEQ ID NO.:84;
(d) a light chain CDR1 of SEQ ID NO. :86;
(e) a light chain CDR2 of SEQ ID NO. :28;
(f) a light chain CDR3 of SEQ ID NO. :87.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:96;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO.:99.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:104;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO:105.
-64-In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO.:108;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO:109.
In one aspect, the invention provides an antibody that binds to death receptor 5 (DR5), comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO. :2;
(c) a heavy chain CDR3 of SEQ ID NO. :98;
(d) a light chain CDR1 of SEQ ID NO. :4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO.:97.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain and a variable light chain comprising an amino acid sequence selected from the group of: SEQ ID NO. :7 and SEQ ID NO. :8; SEQ ID NO. :23 and SEQ ID
NO. :24;
SEQ ID NO.:26 and SEQ ID NO. :24; SEQ ID NO. :23 and SEQ ID NO. :29; SEQ ID
NO. :23 and SEQ ID NO.:30; SEQ ID NO.:26 and SEQ ID NO.:31; SEQ ID NO.:26 and SEQ ID
NO.:32;
SEQ ID NO.:26 and SEQ ID NO.:30; SEQ ID NO.:23 and SEQ ID NO.:31; SEQ ID
NO.:82 and SEQ ID NO.:85; SEQ ID NO.:100 and SEQ ID NO.:101; SEQ ID NO.:102 and SEQ ID
NO.:103; SEQ ID NO.:106 and SEQ ID NO.:107; SEQ ID NO. :94 and SEQ ID NO. :95.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :7 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:8.
-65-In one embodiment, the antibody that binds to death receptor 5 (DR5) comprisesa variable heavy chain comprising an amino acid sequence of SEQ ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ ID NO. :24.
In one embodiment, the antibody that binds to death receptor 5 (DR5) comprisesa variable heavy chain comprising an amino acid sequence of SEQ ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ ID NO. :24.
In one embodiment, the antibody that binds to death receptor 5 (DR5) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ ID NO. :29.
In one embodiment, the antibody that binds to death receptor 5 (DR5) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:30.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:31.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:32.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :26 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:30.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :23 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:31.
-66-In one embodiment, the antibody that binds to death receptor 5 (DR5) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :82 and a variable light chain comprising an amino acid sequence of SEQ ID NO. :85.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO.:100 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:101.
In one embodiment, the antibody that binds to death receptor 5 (DR5) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO.:102 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:103.
In one embodiment, the antibody that binds to death receptor 5 (DR5) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO.:106 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:107.
In one embodiment, the antibody that binds to death receptor 5 (DRS) comprises a variable heavy chain comprising an amino acid sequence of SEQ ID NO. :94 and a variable light chain comprising an amino acid sequence of SEQ ID NO. :95.
In another aspect, the antibody that binds to death receptor 5 (DRS) comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID
NO. :7.
In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but the antibody that binds to DRS
comprising that sequence retains the ability to bind to DRS. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO.
:7. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the antibody that binds to DRS comprises the VH sequence in SEQ ID
NO.:7, including post-translational modifications of that sequence.
-67-In another aspect the antibody that binds to death receptor 5 (DR5) a light chain variable domain (VL) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO. :8.
In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an antibody that binds to DR5 comprising that sequence retains the ability to bind to DR5. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID
NO.:8. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the antibody that binds to DRS comprises the VL
sequence in SEQ ID
NO:8, including post-translational modifications of that sequence.
In another aspect, a bispecific antibody that binds to DRS and a second antigen is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above. In one embodiment, the antibody comprises the VH and VL sequences in SEQ ID NO:7 and SEQ ID NO:8, respectively, including post-translational modifications of those sequences. In one embodiment a bispecific antibody is provided that binds to DRS and a second antigen, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above and wherein the antibody has a format as outlined for the DR5-FAP bispecific antibodies in section C
below. In another embodiment said bispecific antibody that binds to DRS and a second antigen is provided which comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above, and which comprises one or more Fc domain modifications as outlined for the DR5-FAP bispecific antibodies in sections D
and E below.
In a further aspect of the invention, an antibody that binds to DRS according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody. In one embodiment said antibody that binds to DRS according to any of the above embodiments is a human antibody.
-68-C. Exemplary formats of bispecific antibodies binding to DR5 and FAP
In one embodiment, a bispecific antibody that binds to DR5 and FAP comprises an antibody fragment, e.g., a Fv, Fab, Fab', scFv,xFab, scFab, diabody, or F(ab')2 fragment. In another embodiment, the antibody comprises a full length antibody, e.g., an intact IgG1 antibody or other antibody class or isotype as defined herein.
The bispecific antibodies according to the invention are at least bivalent and can be trivalent or multivalent e.g. tetravalent or hexavalent.
The bispecific antibody of the invention comprise an Fc domain, at least one Fab fragment comprising an antigen binding site specific for DR5, and at least one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In another embodiment, the bispecific antibody comprises an Fc domain, at least one Fab fragment comprising an antigen binding site specific for DR5, and at least one Fab fragment comprising an antigen binding site specific for FAP, wherein at least one of the Fab fragments is connected to the first or second subunit of the Fc domain via the light chain (VLCL) and at least one Fab fragment is connected to the first or second subunit of the Fc domain via the heavy chain (VHCH1).
In any of the embodiments, the Fab fragments may be fused to the Fc domain or to each other directly or through a peptide linker, comprising one or more amino acids, typically about 2-20 amino acids. Peptide linkers are known in the art and are described herein.
Suitable, non-immunogenic peptide linkers include, for example, (G4S)., (SG4)., (G4S). or G4(SG4). peptide linkers. "n" is generally a number between 1 and 10, typically between 2 and 4. A particularly suitable peptide linker for fusing the Fab light chains of the first and the second antigen binding moiety to each other is (G4S)2. An exemplary peptide linker suitable for connecting the Fab heavy chains of the first and the second antigen binding moiety is EPKSC(D)-(G4S)2 (SEQ ID
NO.: 318). Additionally, linkers may comprise (a portion of) an immunoglobulin hinge region.
Particularly where an antigen binding moiety is fused to the N-terminus of an Fc domain subunit, it may be fused via an immunoglobulin hinge region or a portion thereof, with or without an additional peptide linker.
Preferably said bispecific antibodies are tetravalent with two binding sites each targeting FAP and DR5, respectively (2+2 format). In another embodiment said bispecific antibodies are tetravalent with three binding sites for DRS and one binding site for FAP (3+1 format). The 3+1 format can be achieved, for example, through fusing one Fab fragment targeting FAP and one
-69-Fab fragment targeting DR5 to the C-terminus of the heavy chain of an IgG
molecule that has two DRS binding sites. This is outlined in more detail below.
In another preferred embodiment said bispecific antibodies are trivalent (2+1 format) with two binding sites each targeting DRS and one binding site targeting FAP. The 2+1 format can be achieved, for example, through fusing a Fab fragment targeting FAP to the C-terminus of the heavy chain of an IgG molecule that has two DRS binding sites., wherein the Fc part of the first antibody is modified according to the knobs-into hole strategy as outlined below.
In another preferred embodiment said bispecific antibodies are bivalent (1+1 format), i.e.
monovalent for each DRS and FAP. Bivalent antibodies of the invention have one binding site targeting DRS and one binding site targeting FAP. The 1+1 format can be achieved, for example, by the Crossmab technology described in Schaefer et al. Proc Natl Acad Sci USA
2011;
108:11187-92 and as outlined below.
Exemplary formats of bispecific antibodies of the invention are given in Figure 25 and 28.
Provided therein are different bispecific antibody formats that are binding to DRS and FAP comprising any of the sequences according to any of the above embodiments.
1. Bispecific DR5-FAP antibodies in a 2+2 Format In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
Since the above bispecific antibody is bivalent both for FAP and DRS, with 2 binding sites each for FAP and DRS, this format is also referred to as "2+2" format.
Exemplary structures of bispecific antibodies with a 2+2 format are depicted in Figure 28. Due to the exchange of either the variable regions or the constant regions, the Fab fragments above are also referred to as "cross-Fab fragment" or "xFab fragment" or "crossover Fab fragment".
In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises an Fc domain,
-70-two Fab fragments comprising each an antigen binding site specific for DR5, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of both Fab fragments comprising an antigen binding site specific for FAP are exchanged.
In another embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of both Fab fragments comprising an antigen binding site specific for DRS are exchanged.
In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein the variable regions of the heavy and light chain of both Fab fragments comprising an antigen binding site specific for FAP are exchanged.
In another embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein the variable regions of the heavy and light chain of both Fab fragments comprising an antigen binding site specific for DRS are exchanged.
Due to the exchange of the variable regions of the Fab heavy and light chain the crossover Fab fragments specific for FAP each comprise a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1), and a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL).
-71-These crossover Fab fragments are also referred to as CrossFab (vLvH ) and each comprise a VLCH1 and a VHCL chain.
In one preferred embodiment a bispecific antibody that binds to DR5 and FAP
according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein the constant regions of the heavy and light chain of both Fab fragments comprising an antigen binding site specific for FAP are exchanged.
In another embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and two Fab fragments comprising each an antigen binding site specific for FAP, wherein the constant regions of the heavy and light chain of both Fab fragments comprising an antigen binding site specific for DRS are exchanged.
Due to the exchange of the constant regions of the Fab heavy and light chain the crossover Fab fragments specific for FAP each comprise a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL), and a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1).
These crossover Fab fragments are also referred to as CrossFab (crou and comprise a VHCL and a VLCH1 chain.
In one embodiment said bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain to which two Fab fragments are fused to the N-terminus and two Fab fragments are fused to the C-terminus, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
In one embodiment two Fab fragments are fused to the N-terminus of the Fc domain through an immunoglobulin hinge region. In one embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In one embodiment the two Fab fragments comprising an antigen binding site specific for DRS and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more
-72-particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin.
In another embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
In one embodiment two Fab fragments comprising the antigen binding site specific for FAP are connected to the Fc domain via a peptide linker. In one embodiment two Fab fragments comprising an antigen binding site specific for FAP are connected to the C-terminus of the first or second subunit of the Fc domain via a peptide linker. In one such embodiment said Fab fragments comprising an antigen binding site specific for FAP are connected to the C-terminus of the second subunit (CH3 chain) of the Fc domain via a peptide linker.
In one embodiment said bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
(i.e. two Fab fragments specific for DRS) and b) two Fab fragments specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
In another embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS wherein either the variable regions or the constant regions of the heavy and light chain are exchanged and b) two Fab fragments specific for FAP.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) two Fab fragments specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises
-73-a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DR5 and b) two Fab fragments specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) two Fab fragments specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule.
In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain of said IgG molecule. In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain of said IgG molecule.
In one embodiment said two Fab fragments are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) two Fab fragments specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule.
In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain of said IgG molecule. In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain of said IgG molecule. In one embodiment said two Fab fragments are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and
-74-b) two Fab fragments specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule.
In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain of said IgG molecule. In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain of said IgG molecule. In one embodiment said two Fab fragments are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In a further preferred embodiment, the two Fab fragments specific for FAP are fused to the IgG molecule by a peptide linker, preferably a peptide linker having a length of about 10 ¨ 30 amino acids. Preferably said peptide linker is a (G4S)2 or (G4S)4 linker.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) two Fab fragments specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain of said IgG molecule by a peptide linker. In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain of said IgG molecule by a peptide linker.
In one embodiment said two Fab fragments are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) two Fab fragments specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain of said IgG molecule by a peptide linker.
-75 -In one embodiment said two Fab fragments are fused to the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain of said IgG
molecule by a peptide linker.
In one embodiment said two Fab fragments are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule by a peptide linker.
In one embodiment, the bispecific antibody comprises an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DR5, and at least one Fab fragment comprising the antigen binding site specific for FAP, wherein at least one of the Fab fragments is connected to the first or second subunit of the Fc domain via the light chain (VLCL) and at least one Fab fragment is connected to the first or second subunit of the Fc domain via the heavy chain (VHCH1).
In one embodiment, the bispecific antibody comprises an Fc domain, two Fab fragments comprising the antigen binding site specific for DR5, and two Fab fragments comprising the antigen binding site specific for FAP, wherein at least one of the Fab fragments is fused to the first or second subunit of the Fc domain via the light chain (VLCL) and at least one Fab fragment is connected to the first or second subunit of the Fc domain via the heavy chain (VHCH1).
In one embodiment, the bispecific antibody comprises a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DR5, wherein said Fab fragments are connected at the C-terminus of the constant light chain (CL) to the first or second subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain.
In one embodiment, the bispecific antibody comprises a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DRS,
-76-wherein said Fab fragments are connected at the C-terminus of the constant light chain (CL) to the N-terminus of the first subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the second subunit (CH3) of the Fc domain.
In one embodiment, said two Fab fragments comprising an antigen binding site specific for DR5 are each fused to the Fc domain through an immunoglobulin hinge region. In a specific embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In one embodiment the two Fab fragments comprising an antigen binding site specific for DRS and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
In one embodiment two Fab fragments comprising the antigen binding site specific for FAP are connected to the Fc domain via a peptide linker.
In one embodiment, the bispecific antibody comprises a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DRS, wherein said Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the N-terminus of the constant light chain (CL) to the first or second subunit of the Fc domain.
In one embodiment, the bispecific antibody comprises a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DRS, wherein said Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the N- terminus of the first subunit of the Fc domain,
-77-c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the N-terminus of the constant light chain (CL) to the N-terminus of the second subunit of the Fc domain.
In one embodiment, said two Fab fragments comprising an antigen binding site specific for DR5 are each fused to the Fc domain through an immunoglobulin hinge region. In a specific embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In one embodiment the two Fab fragments comprising an antigen binding site specific for DRS and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
Exemplary antibodies with a 2+2 format In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a heavy chain CDR1 of SEQ ID NO.:1;
a heavy chain CDR2 of SEQ ID NO. :2;
a heavy chain CDR3 of SEQ ID NO. :3;
a light chain CDR1 of SEQ ID NO. :4;
a light chain CDR2 of SEQ ID NO.:5;
a light chain CDR3 of SEQ ID NO. :6; and b) two Fab fragments specific for FAP, comprising a heavy chain CDR1 of SEQ ID NO. :9;
a heavy chain CDR2 of SEQ ID NO.:10;
a heavy chain CDR3 of SEQ ID NO.:11;
a light chain CDR1 of SEQ ID NO.:12;
a light chain CDR2 of SEQ ID NO.:13;
a light chain CDR3 of SEQ ID NO.:14;
-78-wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a heavy chain CDR1 of SEQ ID NO.:1;
a heavy chain CDR2 of SEQ ID NO. :2;
a heavy chain CDR3 of SEQ ID NO. :3;
a light chain CDR1 of SEQ ID NO. :4;
a light chain CDR2 of SEQ ID NO.:5;
a light chain CDR3 of SEQ ID NO. :6; and b) two Fab fragments specific for FAP, comprising a heavy chain CDR1 of SEQ ID NO. :9;
a heavy chain CDR2 of SEQ ID NO.:10;
a heavy chain CDR3 of SEQ ID NO.:11;
a light chain CDR1 of SEQ ID NO.:12;
a light chain CDR2 of SEQ ID NO.:13;
a light chain CDR3 of SEQ ID NO.:14;
wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8; and
-79-b) two Fab fragments specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16; wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8; and b) two Fab fragments specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16; wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In another embodiment said bispecific antibody of the invention comprises a modification in the Fc part of the IgG molecule, as outlined below.
In one embodiment a bispecific antibody with a 2+2 format as described above is provided comprising two VH (DR5)_Fc part ¨ VH (FAP) -CL chains of SEQ ID
NO.:131, two VL
(DRS)-kappa light chains of SEQ ID NO.:132 and two VLCH1 (FAP) chains of SEQ
ID
NO. :124.
In one embodiment a bispecific antibody with a 2+2 format as described above is provided comprising two VH (DR5)_Fc part ¨ VH (FAP) -CL chains of SEQ ID
NO.:133, two VL
(DRS)-kappa light chains of SEQ ID NO.:132 and two VLCH1 (FAP) chains of SEQ
ID
NO. :124.
In one embodiment a bispecific antibody with a 2+2 format as described above is provided comprising two VH (DR5)_Fc part ¨ VH (FAP) -CL chains of SEQ ID
NO.:134, two VL
(DRS)-kappa light chains of SEQ ID NO.:132 and two VLCH1 (FAP) chains of SEQ
ID
NO. :124.
In one embodiment a bispecific antibody with a 2+2 format as described above is provided comprising two VL (DR5)_CH1- Fc part-VH(FAn_CH1 chains of SEQ ID NO.
135, two
-80-VH(DR5) ¨CL chains of SEQ ID NO.:136 and two VL (FAP) ¨kappa light chains of SEQ ID
NO.:137.
In one embodiment a bispecific antibody with a 2+2 format as described above is provided, comprising two VH (DRS) CL- Fc- peptide linker-VH(FAn -CH1 chains of SEQ ID
NO.:138, two VL(DRS) ¨CH1 chains of SEQ ID NO.:139 and two VL (FAP) ¨kappa light chains of SEQ ID NO.:137.
2. Bispecific DR5-FAP antibodies in a 2+1 Format In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising an antigen binding site specific for DR5, and one Fab fragment comprising the antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
Since the above bispecific antibodies are trivalent with one binding site for FAP and two binding sites for DR5, this format is also referred to as "2+1" format. Hence the bispecific antibodies provided in this section are bivalent for DR5 and monovalent for FAP.
An exemplary structure of a bispecific antibodies with a 2+1 format are depicted in Figure 25 a) and b). Due to the exchange of either the variable regions or the constant regions, the Fab fragments above are also referred to as "cross-Fab fragment" or "xFab fragment" or "crossover Fab fragment".
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of the Fab fragments comprising an antigen binding site specific for FAP are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises
-81 -an Fc domain, two Fab fragments comprising each an antigen binding site specific for DR5, and one Fab fragment comprising an antigen binding site specific for FAP, wherein the variable regions of the heavy and light chain of the Fab fragment comprising an antigen binding site specific for FAP are exchanged.
Due to the exchange of the variable regions of the Fab heavy and light chain the crossover Fab fragment specific for FAP each comprise a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1), and a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL).
These crossover Fab fragment is also referred to as CrossFab (VH) and comprises a VLCH1 and a VHCL chain.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, two Fab fragments comprising each an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein the constant regions of the heavy and light chain of both Fab fragments comprising the antigen binding site specific for FAP are exchanged.
Due to the exchange of the constant regions of the Fab heavy and light chain the crossover Fab fragment specific for FAP each comprise a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL), and a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1).
These crossover Fab fragments is also referred to as CrossFab (CLCH 1 ) and comprises a VHCL and a VLCH1 chain.
In one embodiment said bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain to which two Fab fragments are fused to the N-terminus.
In one embodiment two Fab fragments are fused to the N-terminus of the Fc domain through an immunoglobulin hinge region. In one embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human
-82-immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
Bispecific DR5-FAP antibodies in a 2+1 Format with FAP binder fused to C-terminus In one embodiment the two Fab fragments comprising an antigen binding site specific for DR5 and the Fc domain are part of an immunoglobulin molecule. In one embodiment one Fab fragment comprising the antigen binding site specific for FAP is fused to the C-terminus of the first or second subunit of the Fc domain via a peptide linker. In one such embodiment said Fab fragment comprising an antigen binding site specific for FAP is fused to the C-terminus of the second subunit (CH3 chain) of the Fc domain via a peptide linker. An exemplary structure of a bispecific DR5-FAP antibody in a 2+1 Format with the FAP binder fused to C-terminus is depicted in Figure 25 a).
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DR5 and b) one Fab fragment specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) one Fab fragment specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) one Fab fragment specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged.
-83-In one preferred embodiment a bispecific antibody that binds to DR5 and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) one Fab fragment specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged, wherein the Fab fragment is fused to the constant heavy chain of said IgG molecule.
In one embodiment said Fab fragment specific for FAP of b) is fused to the C-terminus of the first or second subunit of the Fc domain of said IgG molecule. In one embodiment said Fab fragment of b) is fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) one Fab fragment specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the Fab fragment is fused to the constant heavy chain of said IgG molecule.
In one embodiment said Fab fragment specific for FAP of b) is fused to the C-terminus of the first or second subunit of the Fc domain of said IgG molecule. In one embodiment said Fab fragment of b) is fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS
and b) one Fab fragment specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the Fab fragment is fused to the constant heavy chain of said IgG molecule.
In one embodiment said Fab fragment specific for FAP of b) is fused to the C-terminus of the first or second subunit of the Fc domain of said IgG molecule. In one embodiment said Fab
-84-fragment of b) is fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
Bispecific DR5-FAP antibodies in a 2+1 Format with FAP binder fused to the N-terminus In another embodiment one Fab fragment comprising an antigen binding site specific for DR5, one Fab fragment comprising an antigen binding site specific for FAP and the Fc domain are part of an immunoglobulin molecule. In one embodiment another Fab fragment comprising an antigen binding site specific for DRS is fused to the N-terminus of the Fab fragment comprising an antigen binding site specific for DRS of the IgG molecule via a peptide linker. An exemplary structure of a bispecific DRS-FAP antibody in a 2+1 Format with the FAP binder fused to N-terminus is depicted in Figure 25 b).
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with one binding site (Fab fragment) specific for DRS and one binding site (Fab fragment) specific for FAP, wherein the Fab fragment specific for FAP is a Crossfab fragment (i.e. either the variable regions or the constant regions of the heavy and light chain are exchanged).
b) one Fab fragment specific for DRS.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with one binding site specific for DRS
and one binding site specific for FAP, wherein the Fab fragment specific for FAP is a CrossFab (NH) fragment (i.e. the variable regions of the heavy and light chain are exchanged).
b) one Fab fragment specific for DRS.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with one binding site specific for DRS
and one binding site specific for FAP, wherein the Fab fragment specific for FAP is a CrossFah (cLcm fragment (i.e. the constant regions of the heavy and light chain are exchanged).
b) one Fab fragment specific for DRS.
-85-In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with one binding site specific for DR5 and one binding site specific for FAP, wherein the Fab fragment specific for FAP is a Crossfab fragment (i.e. either the variable regions or the constant regions of the heavy and light chain are exchanged).
b) one Fab fragment specific for DR5, wherein said Fab fragment is fused to the N-terminus of the variable heavy or light chain of the IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with one binding site (Fab fragment) specific for DRS and one binding site specific for FAP, wherein the Fab fragment specific for FAP is a CrossFab (vLvH ) fragment (i.e. the variable regions of the heavy and light chain are exchanged).
b) one Fab fragment specific for DRS, wherein said Fab fragment is fused to the N-terminus of the variable heavy or light chain of the IgG molecule.
In one embodiment, the Fab fragment specific for DRS of b) is fused to the N-terminus of the variable heavy or light chain of the Fab fragment specific for DRS of a).
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with one binding site specific for DRS
and one binding site specific for FAP, wherein the Fab fragment specific for FAP is a CrossFah (cLcm fragment (i.e. the constant regions of the heavy and light chain are exchanged).
b) one Fab fragment specific for DRS, wherein said Fab fragment is fused to the N-terminus of the variable heavy or light chain of the IgG molecule In one embodiment, the Fab fragment specific for DRS of b) is fused to the N-terminus of the variable heavy or light chain of the Fab fragment specific for DRS of a).
-86-In a further preferred embodiment, the Fab fragment specific for DR5 is fused to the IgG
molecule by a peptide linker, preferably a peptide linker having a length of about 10 ¨ 30 amino acids. Preferably said peptide linker is a (G4S)2 or (G4S)4 linker.
In another embodiment said bispecific antibody of the invention comprises a modification in the Fc part of the IgG molecule, as outlined below.
3. Bispecific DR5-FAP antibodies in a 3+1 Format In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, three Fab fragments comprising each an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
Since the above bispecific antibody is tetravalent with one binding site for FAP and three binding sites for DRS, this format is also referred to as "3+1" format. Hence the bispecific molecules described in this section are trivalent for DRS and monovalent for FAP.
An exemplary structure of a bispecific antibody with a 3 +1 format is depicted in Figure 25 c). Due to the exchange of either the variable regions or the constant regions, the Fab fragments above are also referred to as "cross-Fab fragment" or "xFab fragment" or "crossover Fab fragment".
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, three Fab fragments comprising each an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of the Fab fragments comprising an antigen binding site specific for FAP are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises
-87-an Fc domain, three Fab fragments comprising each an antigen binding site specific for DR5, and one Fab fragment comprising an antigen binding site specific for FAP, wherein the variable regions of the heavy and light chain of the Fab fragment comprising an antigen binding site specific for FAP are exchanged.
Due to the exchange of the variable regions of the Fab heavy and light chain the crossover Fab fragment specific for FAP comprises a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1), and a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL). This crossover Fab fragments is also referred to as CrossFab (VLvH ) and comprises a VLCH1 and a VHCL chain.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, three Fab fragments comprising each an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein the constant regions of the heavy and light chain of the Fab fragment comprising the antigen binding site specific for FAP are exchanged.
Due to the exchange of the constant regions of the Fab heavy and light chain the crossover Fab fragment specific for FAP each comprise a peptide chain composed of the heavy chain variable region (VH) and the light chain constant region (CL), and a peptide chain composed of the light chain variable region (VL) and the heavy chain constant region (CH1).
This crossover Fab fragment is also referred to as CrossFab (crcHo and comprise a VHCL and a VLCH1 chain.
In one embodiment said bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain to which two Fab fragments are fused to the N terminus and two Fab fragments are fused to the C-terminus, wherein either the variable regions or the constant regions of the heavy and light chain of the one Fab fragment specific fro FAP are exchanged.
In one embodiment two Fab fragments are fused to the N-terminus of the Fc domain through an immunoglobulin hinge region. In one embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In one embodiment the two Fab fragments comprising an antigen binding site specific for DRS and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin.
In another
-88-embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS and b) one Fab fragment specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
c) one Fab fragment specific for DRS.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS and b) one Fab fragment specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged.
c) one Fab fragment specific for DRS.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS and b) one Fab fragment specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged.
c) one Fab fragment specific for DRS.
In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS and
-89-b) one Fab fragment specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
c) one Fab fragment specific for DR5, wherein the Fab fragments of b) and c) are fused to the C-terminus of the first or second subunit of the Fc domain of said IgG molecule.
In one embodiment said two Fab fragments of b) and c) are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS and b) one Fab fragment specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged.
c) one Fab fragment specific for DRS, wherein the Fab fragments of b) and c) are fused to the first or second subunit of the Fc domain of said IgG molecule.
In one embodiment said two Fab fragments of b) and c) are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites (Fab fragments) specific for DRS and b) one Fab fragment specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged.
c) one Fab fragment specific for DRS, wherein the Fab fragments of b) and c) are fused to the first or second subunit of the Fc domain of said IgG molecule.
In one embodiment said two Fab fragments of b) and c) are fused to the C-terminus of the second subunit (CH3) of the Fc domain of said IgG molecule.
-90-In a further preferred embodiment, the two Fab fragments of b) and c) of any of the embodiments described in this section are fused to the IgG molecule by a peptide linker, preferably a peptide linker having a length of about 10 ¨ 30 amino acids.
Preferably said peptide linker is a (G4S)2 or (G4S)4 linker.
In another embodiment said bispecific antibody of the invention comprises a modification in the Fc part of the IgG molecule, as outlined below.
4. Bispecific DR5-FAP antibodies in a 1+1 Format In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises an Fc domain, one Fab fragment comprising an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
Since the above bispecific antibody is bivalent with one binding site for FAP
and one binding site for DRS, this format is also referred to as "1+1" format. Hence the bispecific antibodies described in this section are monovalent for DRS and monovalent for FAP. An exemplary structure of a bispecific antibody with a 1 +1 format is depicted in Figure 25 d). Due to the exchange of either the variable regions or the constant regions, the Fab fragment above is also referred to as "cross-Fab fragment" or "xFab fragment" or "crossover Fab fragment". The IgG molecule in a 1+1 format is also referred to as Crossmab format (see Schaefer et al. Proc Natl Acad Sci USA 2011; 108:11187-92).
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, one Fab fragment comprising an antigen binding site specific for DRS, wherein either the variable regions or the constant regions of the heavy and light chain of the Fab fragment comprising an antigen binding site specific for DRS are exchanged.
and one Fab fragment comprising an antigen binding site specific for FAP.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises
-91-an Fc domain, one Fab fragment comprising an antigen binding site specific for DR5, and one Fab fragment comprising an antigen binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of the Fab fragment comprising an antigen binding site specific for FAP are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, one Fab fragment comprising an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein the variable regions of the heavy and light chain of the Fab fragment comprising an antigen binding site specific for FAP are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain, one Fab fragments comprising an antigen binding site specific for DRS, and one Fab fragment comprising an antigen binding site specific for FAP, wherein the constant regions of the heavy and light chain of the Fab fragment comprising the antigen binding site specific for FAP are exchanged.
In one embodiment said bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Fc domain to which two Fab fragments are fused to the N-terminus, wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged. In one embodiment the two Fab fragments are fused to the N-terminus of the Fc domain through an immunoglobulin hinge region. In one embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In one embodiment the Fab fragment comprising an antigen binding site specific for DRS, the Fab fragment comprising an antigen binding site specific for FAP and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG
class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment the immunoglobulin is an IgG4 subclass
-92-immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises an Immunoglobulin G (IgG) molecule with one binding site specific for DRS and one binding site specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain of one arm (Fab fragment) of the IgG molecule are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Immunoglobulin G (IgG) molecule with one binding site specific for DRS and one binding site specific for FAP, wherein the variable regions of the heavy and light chain of one arm (Fab fragment) of the IgG molecule are exchanged.
This antibody format is also referred to as CrossMab(vHvL).
In one embodiment the variable regions of the heavy and light chain of the one arm (Fab fragment) of the IgG molecule which comprises the binding site specific for FAP are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Immunoglobulin G (IgG) molecule with one binding site specific for DRS and one binding site specific for FAP, wherein the constant regions of the heavy and light chain of one arm (Fab fragment) of the IgG molecule are exchanged. This antibody format is also referred to as CrossMab(cm cL) In one embodiment the constant regions of the heavy and light chain of the one arm (Fab fragment) of the IgG molecule which comprises the binding site specific for FAP are exchanged.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises an Immunoglobulin G (IgG) molecule with one binding site specific for DRS and one binding site specific for FAP, wherein the complete VH-CH1 and VL-CL domains of one arm (Fab fragment) of the IgG molecule are exchanged. This means that at least one of the Fab fragments is fused to the N-terminus of the Fc domain via the light chain (VLCL). In one embodiment the other Fab fragment is fused to the the N-terminus of the Fc domain via the heavy chain (VHCH1).
-93-This antibody format is also referred to as CrossMabFab. In one embodiment both Fab fragments are are fused to the N-terminus of the Fc domain through an immunoglobulin hinge region.
D. Fc domain modifications reducing Fc receptor binding and/or effector function In one preferred embodiment a bispecific antibody that binds to DR5 and FAP
according to any of the above embodiments comprises an Immunoglobulin G (IgG) molecule wherein the Fc part is modified. The modified Fc part has a reduced binding affinity for the Fcy receptors compared to a wildtype Fc part.
The Fc domain of the bispecific antibodies of the invention consists of a pair of polypeptide chains comprising heavy chain domains of an immunoglobulin molecule. For example, the Fc domain of an immunoglobulin G (IgG) molecule is a dimer, each subunit of which comprises the CH2 and CH3 IgG heavy chain constant domains. The two subunits of the Fc domain are capable of stable association with each other.
In one embodiment according the invention the Fc domain of the bispecific antibodies of the invention is an IgG Fc domain. In a particular embodiment the Fc domain is an IgGi Fc domain.
In another embodiment the Fc domain is an IgG4 Fc domain. In a more specific embodiment, the Fc domain is an IgG4 Fc domain comprising an amino acid substitution at position S228 (Kabat numbering), particularly the amino acid substitution S228P. In a more specific embodiment, the Fc domain is an IgG4 Fc domain comprising amino acid substitutions L235E and S228P and P329G. This amino acid substitution reduces in vivo Fab arm exchange of IgG4 antibodies (see Stubenrauch et al., Drug Metabolism and Disposition 38, 84-91 (2010)). In a further particular embodiment the Fc domain is human. An exemplary sequence of a human IgGi Fc region is given in SEQ ID NO.:151.
The Fc domain confers favorable pharmacokinetic properties to the bispecific antibodies of the invention, including a long serum half-life which contributes to good accumulation in the target tissue and a favorable tissue-blood distribution ratio. At the same time it may, however, lead to undesirable targeting of the bispecific antibodies of the invention to cells expressing Fc receptors rather than to the preferred antigen-bearing cells. Accordingly, in particular embodiments the Fc domain of the the bispecific antibodies of the invention exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgGi Fc domain. In one such embodiment the Fc domain (or the bispecific antibodies of the invention comprising said Fc domain) exhibits less than 50%, preferably less than 20%, more preferably less than 10% and
-94-most preferably less than 5% of the binding affinity to an Fc receptor, as compared to a native IgGi Fc domain (or a bispecific antibodies of the invention comprising a native IgGi Fc domain), and/or less than 50%, preferably less than 20%, more preferably less than 10%
and most preferably less than 5% of the effector function, as compared to a native IgGi Fc domain domain (or a bispecific antibodies of the invention comprising a native IgGi Fc domain). In one embodiment, the Fc domain (or the bispecific antibodies of the invention comprising said Fc domain) does not substantially bind to an Fc receptor and/or induce effector function. In a particular embodiment the Fc receptor is an Fcy receptor. In one embodiment the Fc receptor is a human Fc receptor. In one embodiment the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fcy receptor, more specifically human Fc7RIIIa, FcyRI or Fc7RIIa, most specifically human Fc7RIIIa. In one embodiment the Fc receptor is an inhibitory Fc receptor. In a specific embodiment the Fc receptor is an inhibitory human Fcy receptor, more specifically human FcgRIIB. In one embodiment the effector function is one or more of CDC, ADCC, ADCP, and cytokine secretion. In a particular embodiment the effector function is ADCC. In one embodiment the Fc domain domain exhibits substantially similar binding affinity to neonatal Fc receptor (FcRn), as compared to a native IgGi Fc domain domain. Substantially similar binding to FcRn is achieved when the Fc domain (or the bispecific antibodies of the invention comprising said Fc domain) exhibits greater than about 70%, particularly greater than about 80%, more particularly greater than about 90%
of the binding affinity of a native IgGi Fc domain (or the bispecific antibodies of the invention comprising a native IgGi Fc domain) to FcRn.
In certain embodiments the Fc domain is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain. In particular embodiments, the Fc domain of the bispecific antibodies of the invention comprises one or more amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function. Typically, the same one or more amino acid mutation is present in each of the two subunits of the Fc domain. In one embodiment the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor. In one embodiment the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor by at least 2-fold, at least 5-fold, or at least 10-fold. In embodiments where there is more than one amino acid mutation that reduces the binding affinity of the Fc domain to the Fc receptor, the combination of these amino acid mutations may reduce the binding affinity of the Fc domain to an Fc receptor by at least 10-fold, at least 20-fold, or even at least 50-fold. In one embodiment the bispecific
-95-antibodies of the invention comprising an engineered Fc domain exhibits less than 20%, particularly less than 10%, more particularly less than 5% of the binding affinity to an Fc receptor as compared to a bispecific antibodies of the invention comprising a non-engineered Fc domain. In a particular embodiment the Fc receptor is an Fcy receptor. In some embodiments the Fc receptor is a human Fc receptor. In one embodiment the Fc receptor is an inhibitory Fc receptor. In a specific embodiment the Fc receptor is an inhibitory human Fcy receptor, more specifically human FcgRIIB. In some embodiments the Fc receptor is an activating Fc receptor.
In a specific embodiment the Fc receptor is an activating human Fcy receptor, more specifically human FcyRIIIa, FcyRI or Fc7RIIa, most specifically human FcyRIIIa.
Preferably, binding to each of these receptors is reduced. In some embodiments binding affinity to a complement component, specifically binding affinity to C 1 q, is also reduced. In one embodiment binding affinity to neonatal Fc receptor (FcRn) is not reduced. Substantially similar binding to FcRn, i.e.
preservation of the binding affinity of the Fc domain to said receptor, is achieved when the Fc domain (or the bispecific antibodies of the invention comprising said Fc domain) exhibits greater than about 70% of the binding affinity of a non-engineered form of the Fc domain (or the bispecific antibodies of the invention comprising said non-engineered form of the Fc domain) to FcRn. The Fc domain, or the bispecific antibodies of the invention of the invention comprising said Fc domain, may exhibit greater than about 80% and even greater than about 90% of such affinity. In certain embodiments the Fc domain of the bispecific antibodies of the invention is engineered to have reduced effector function, as compared to a non-engineered Fc domain. The reduced effector function can include, but is not limited to, one or more of the following:
reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen-presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced dendritic cell maturation, or reduced T cell priming. In one embodiment the reduced effector function is one or more of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In a particular embodiment the reduced effector function is reduced ADCC. In one embodiment the reduced ADCC is less than 20% of the ADCC induced by a non-engineered Fc domain (or a bispecific antibody of the invention comprising a non-engineered Fc domain).
-96-In one embodiment the amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function is an amino acid substitution. In one embodiment the Fc domain comprises an amino acid substitution at a position of E233, L234, L235, N297, P331 and P329. In a more specific embodiment the Fc domain comprises an amino acid substitution at a position of L234, L235 and P329. In some embodiments the Fc domain comprises the amino acid substitutions L234A and L235A. In one such embodiment, the Fc domain is an IgGi Fc domain, particularly a human IgGi Fc domain. In one embodiment the Fc domain comprises an amino acid substitution at position P329. In a more specific embodiment the amino acid substitution is P329A or P329G, particularly P329G. In one embodiment the Fc domain comprises an amino acid substitution at position P329 and a further amino acid substitution at a position selected from E233, L234, L235, N297 and P331. In a more specific embodiment the further amino acid substitution is E233P, L234A, L235A, L235E, N297A, N297D or P331S. In particular embodiments the Fc domain comprises amino acid substitutions at positions P329, L234 and L235. In more particular embodiments the Fc domain comprises the amino acid mutations L234A, L235A and P329G ("P329G LALA"). In one such embodiment, the Fc domain is an IgGi Fc domain, particularly a human IgGi Fc domain. The "P329G
LALA"
combination of amino acid substitutions almost completely abolishes Fcy receptor binding of a human IgGi Fc domain, as described in PCT patent application no.
PCT/EP2012/055393, incorporated herein by reference in its entirety. PCT/EP2012/055393 also describes methods of preparing such mutant Fc domains and methods for determining its properties such as Fc receptor binding or effector functions.
IgG4 antibodies exhibit reduced binding affinity to Fc receptors and reduced effector functions as compared to IgGi antibodies. Hence, in some embodiments the Fc domain of the bispecific antibodies of the invention is an IgG4 Fc domain, particularly a human IgG4 Fc domain. In one embodiment the IgG4 Fc domain comprises amino acid substitutions at position S228, specifically the amino acid substitution S228P. To further reduce its binding affinity to an Fc receptor and/or its effector function, in one embodiment the IgG4 Fc domain comprises an amino acid substitution at position L235, specifically the amino acid substitution L235E. In another embodiment, the IgG4 Fc domain comprises an amino acid substitution at position P329, specifically the amino acid substitution P329G. In a particular embodiment, the IgG4 Fc domain comprises amino acid substitutions at positions S228, L235 and P329, specifically amino acid substitutions S228P, L235E and P329G. Such IgG4 Fc domain mutants and their Fcy receptor
-97-binding properties are described in PCT patent application no.
PCT/EP2012/055393, incorporated herein by reference in its entirety.
In a particular embodiment the Fc domain exhibiting reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgGi Fc domain, is a human IgGi Fc domain comprising the amino acid substitutions L234A, L235A and optionally P329G, or a human IgG4 Fc domain comprising the amino acid substitutions S228P, L235E and optionally P329G.
In certain embodiments N-glycosylation of the Fc domain has been eliminated.
In one such embodiment the Fc domain comprises an amino acid mutation at position N297, particularly an amino acid substitution replacing asparagine by alanine (N297A) or aspartic acid (N297D).
In addition to the Fc domains described hereinabove and in PCT patent application no.
PCT/EP2012/055393, Fc domains with reduced Fc receptor binding and/or effector function also include those with substitution of one or more of Fc domain residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US
Patent No.
7,332,581).
Mutant Fc domains can be prepared by amino acid deletion, substitution, insertion or modification using genetic or chemical methods well known in the art. Genetic methods may include site-specific mutagenesis of the encoding DNA sequence, PCR, gene synthesis, and the like. The correct nucleotide changes can be verified for example by sequencing.
Binding to Fc receptors can be easily determined e.g. by ELISA, or by Surface Plasmon Resonance (SPR) using standard instrumentation such as a BIAcore instrument (GE Healthcare), and Fc receptors such as may be obtained by recombinant expression. A suitable such binding assay is described herein. Alternatively, binding affinity of Fc domains or cell activating bispecific antigen binding molecules comprising an Fc domain for Fc receptors may be evaluated using cell lines known to express particular Fc receptors, such as human NK
cells expressing Fc7IIIa receptor.
Effector function of an Fc domain, or bispecific antibodies of the invention comprising an Fc domain, can be measured by methods known in the art. A suitable assay for measuring ADCC is described herein. Other examples of in vitro assays to assess ADCC activity of a molecule of interest are described in U.S. Patent No. 5,500,362; Hellstrom et al. Proc Nati Acad Sci USA 83, 7059-7063 (1986) and Hellstrom et al., Proc Nati Acad Sci USA 82, 1499-1502 (1985); U.S.
-98-Patent No. 5,821,337; Bruggemann et al., J Exp Med 166, 1351-1361 (1987).
Alternatively, non-radioactive assays methods may be employed (see, for example, ACTITm non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA); and CytoTox 96 non-radioactive cytotoxicity assay (Promega, Madison, WI)). Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g. in a animal model such as that disclosed in Clynes et al., Proc Natl Acad Sci USA 95, 652-656 (1998).
In some embodiments, binding of the Fc domain to a complement component, specifically to C 1 q, is reduced. Accordingly, in some embodiments wherein the Fc domain is engineered to have reduced effector function, said reduced effector function includes reduced CDC. C 1 q binding assays may be carried out to determine whether the bispecific antibodies of the invention is able to bind C lq and hence has CDC activity. See e.g., C lq and C3c binding ELISA in WO
2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J Immunol Methods 202, 163 (1996);
Cragg et al., Blood 101, 1045-1052 (2003); and Cragg and Glennie, Blood 103, (2004)).
The following section describes preferred embodiments of the bispecific antibodies of the invention comprising Fc domain modifications reducing Fc receptor binding and/or effector function.
In one preferred embodiment a bispecific antibody that binds to DR5 and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DR5, wherein the Fc domain exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain and b) two Fab fragments specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
In one preferred embodiment a bispecific antibody that binds to DR5 and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein the Fc domain comprises one or more amino acid substitution that reduces binding to an Fc receptor and/or effector function.
-99-b) two Fab fragments specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
In one preferred embodiment a bispecific antibody that binds to DR5 and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein said one or more amino acid substitution is at one or more position of L234, L235, and P329 b) two Fab fragments specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G.
b) two Fab fragments specific for FAP, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged.
In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G.
b) two Fab fragments specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged.
In one preferred embodiment a bispecific antibody that binds to DRS and FAP
according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein each subunit of the Fc domain comprises three amino acid substitutions that
-100-reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G.
b) two Fab fragments specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged.
.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a heavy chain CDR1 consisting of SEQ ID NO.:1;
a heavy chain CDR2 of SEQ ID NO.:2;
a heavy chain CDR3 of SEQ ID NO.:3;
-101-a light chain CDR1 of SEQ ID NO. :4;
a light chain CDR2 of SEQ ID NO. :5;
a light chain CDR3 of SEQ ID NO.:6;
wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to activating and inhibitory Fc receptors and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, comprising a heavy chain CDR1 of SEQ ID NO. :9;
a heavy chain CDR2 of SEQ ID NO.:10;
a heavy chain CDR3 of SEQ ID NO.:11;
a light chain CDR1 of SEQ ID NO.:12;
a light chain CDR2 of SEQ ID NO.:13;
a light chain CDR3 of SEQ ID NO.:14;
wherein either the variable regions or the constant regions of the heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG
molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DR5, comprising a heavy chain CDR1 consisting of SEQ ID NO.:1;
a heavy chain CDR2 of SEQ ID NO.:2;
a heavy chain CDR3 of SEQ ID NO.:3;
a light chain CDR1 of SEQ ID NO. :4;
a light chain CDR2 of SEQ ID NO. :5;
a light chain CDR3 of SEQ ID NO.:6;
-102-wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, comprising a heavy chain CDR1 of SEQ ID NO.:9;
a heavy chain CDR2 of SEQ ID NO.:10;
a heavy chain CDR3 of SEQ ID NO.:11;
a light chain CDR1 of SEQ ID NO.:12;
a light chain CDR2 of SEQ ID NO.:13;
a light chain CDR3 of SEQ ID NO.:14;
wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a heavy chain CDR1 consisting of SEQ ID NO.:1;
a heavy chain CDR2 of SEQ ID NO.:2;
a heavy chain CDR3 of SEQ ID NO.:3;
a light chain CDR1 of SEQ ID NO. :4;
a light chain CDR2 of SEQ ID NO. :5;
a light chain CDR3 of SEQ ID NO.:6;
wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, comprising a heavy chain CDR1 of SEQ ID NO.:9;
a heavy chain CDR2 of SEQ ID NO.:10;
a heavy chain CDR3 of SEQ ID NO.:11;
-103-a light chain CDR1 of SEQ ID NO.:12;
a light chain CDR2 of SEQ ID NO.:13;
a light chain CDR3 of SEQ ID NO.:14;
wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8;
wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16, wherein either the variable regions or the constant regions of the heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DRS and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8;
wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region
-104-comprising an amino acid sequence of SEQ ID NO.:16wherein the constant regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises a) an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, comprising a variable heavy chain of SEQ ID NO. :7 and a variable light chain of SEQ ID
NO.:8;
wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G and b) two Fab fragments specific for FAP, comprising a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16, wherein the variable regions of the Fab heavy and light chain are exchanged, wherein the two Fab fragments are fused to the constant heavy chain of said IgG molecule by a peptide linker.
E. Fc domain modifications promoting heterodimerization The bispecific DRS-FAP antibodies of the invention comprise different antigen binding moieties, fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised in two non-identical polypeptide chains.
Recombinant co-expression of these polypeptides and subsequent dimerization leads to several possible combinations of the two polypeptides. To improve the yield and purity of the bispecific antibodies of the invention in recombinant production, it will thus be advantageous to introduce in the Fc domain of the bispecific antibodies of the invention a modification promoting the association of the desired polypeptides.
Accordingly, in particular embodiments the Fc domain of the bispecific antibodies of the invention comprises a modification promoting the association of the first and the second subunit of the Fc domain. The site of most extensive protein-protein interaction between the two subunits of a human IgG Fc domain is in the CH3 domain of the Fc domain. Thus, in one embodiment said modification is in the CH3 domain of the Fc domain.
-105-In a specific embodiment said modification is a so-called "knob-into-hole"
modification, comprising a "knob" modification in one of the two subunits of the Fc domain and a "hole"
modification in the other one of the two subunits of the Fc domain.
The knob-into-hole technology is described e.g. in US 5,731,168; US 7,695,936;
Ridgway et al., Prot Eng 9, 617-621 (1996) and Carter, J Immunol Meth 248, 7-15 (2001).
Generally, the method involves introducing a protuberance ("knob") at the interface of a first polypeptide and a corresponding cavity ("hole") in the interface of a second polypeptide, such that the protuberance can be positioned in the cavity so as to promote heterodimer formation and hinder homodimer formation. Protuberances are constructed by replacing small amino acid side chains from the interface of the first polypeptide with larger side chains (e.g.
tyrosine or tryptophan).
Compensatory cavities of identical or similar size to the protuberances are created in the interface of the second polypeptide by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine).
Accordingly, in a particular embodiment, in the CH3 domain of the first subunit of the Fc domain of the bispecific antibodies of the invention an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.
The protuberance and cavity can be made by altering the nucleic acid encoding the polypeptides, e.g. by site-specific mutagenesis, or by peptide synthesis.
In a specific embodiment, in the CH3 domain of the first subunit of the Fc domain the threonine residue at position 366 is replaced with a tryptophan residue (T366W), and in the CH3 domain of the second subunit of the Fc domain the tyrosine residue at position 407 is replaced with a valine residue (Y407V). In one embodiment, in the second subunit of the Fc domain additionally the threonine residue at position 366 is replaced with a serine residue (T366S) and the leucine residue at position 368 is replaced with an alanine residue (L368A).
In yet a further embodiment, in the first subunit of the Fc domain additionally the serine residue at position 354 is replaced with a cysteine residue (S354C), and in the second subunit of the Fc domain additionally the tyrosine residue at position 349 is replaced by a cysteine residue (Y349C). Introduction of these two cysteine residues results in formation of a disulfide bridge
-106-between the two subunits of the Fc domain, further stabilizing the dimer (Carter, J Immunol Methods 248, 7-15 (2001)).
In an alternative embodiment a modification promoting association of the first and the second subunit of the Fc domain comprises a modification mediating electrostatic steering effects, e.g.
as described in PCT publication WO 2009/089004. Generally, this method involves replacement of one or more amino acid residues at the interface of the two Fc domain subunits by charged amino acid residues so that homodimer formation becomes electrostatically unfavorable but heterodimerization electrostatically favorable.
In one embodiment a bispecific antibody that binds to DR5 and FAP according to any of the above embodiments comprises an Immunoglobulin G (IgG) molecule with two binding sites specific for DRS, wherein the Fc part of the first heavy chain comprises a first dimerization module and the Fc part of the second heavy chain comprises a second dimerization module allowing a heterodimerization of the two heavy chains of the IgG molecule.
In a further preferred embodiment, the first dimerization module comprises knobs and the second dimerization module comprises holes according to the knobs into holes strategy (see Carter P.; Ridgway J.B.B.; Presta L.G.: Immunotechnology, Volume 2, Number 1, February 1996 , pp. 73-73(1)).
F. Nucleic Acid sequences, vectors and methods of The invention further provides isolated polynucleotides encoding a bispecific antibody or an antibody binding to DRS as described herein or a fragment thereof. The polynucleotides encoding bispecific antibodies or the antibodies binding to DRS of the invention may be expressed as a single polynucleotide that encodes the entire bispecific antigen binding molecule or the entire antibody binding to DRS or as multiple (e.g., two or more) polynucleotides that are co-expressed. Polypeptides encoded by polynucleotides that are co-expressed may associate through, e.g., disulfide bonds or other means to form a functional bispecific antibody or an antibody binding to DRS. For example, the light chain portion of a Fab fragment may be encoded by a separate polynucleotide from the portion of the bispecific antibody or the antibody binding to DRS comprising the heavy chain portion of the Fab fragment, an Fc domain subunit and optionally (part of) another Fab fragment. When co-expressed, the heavy chain polypeptides will associate with the light chain polypeptides to form the Fab fragment. In another example, the portion of the bispecific antibody or the antibody binding to DRS provided therein comprising one of the two Fc domain subunits and optionally (part of) one or more Fab fragments could be encoded by a separate polynucleotide from the portion of the bispecific
-107-antibody or the antibody binding to DR5 provided therein comprising the the other of the two Fc domain subunits and optionally (part of) a Fab fragment. When co-expressed, the Fc domain subunits will associate to form the Fc domain.
In one embodiment, the present invention is directed to an isolated polynucleotide encoding a bispecific antibody of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable heavy chain sequence as shown in SEQ ID NOs 167, 175, 183, 191, 199, 207 and 209.
In one embodiment, the present invention is directed to an isolated polynucleotide encoding a bispecific antibody of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable light chain sequence as shown in SEQ ID NOs 171, 179, 187, 195, 203, 208 and 210.
In another embodiment, the present invention is directed to an isolated polynucleotide encoding a bispecific antibody or fragment thereof, wherein the polynucleotide comprises a sequence that encodes a polypeptide sequence as shown in SEQ ID NOs 222, 224, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 258, 259, 260, 261, 264 and 265.
In one embodiment, the present invention is directed to an isolated polynucleotide encoding an antibody binding to DR5 of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable heavy chain sequence as shown in SEQ ID NOs 167, 175, 183, 191 and 199.
In one embodiment, the present invention is directed to an isolated polynucleotide encoding an antibody binding to DR5 of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable light chain sequence as shown in SEQ ID NOs 171, 179, 187, 195 and 203.
In another embodiment, the invention is directed to an isolated polynucleotide encoding a bispecific antibody of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable heavy chain sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs 167, 175, 183, 191, 199, 207 or 209.
In another embodiment, the invention is directed to an isolated polynucleotide encoding a bispecific antibody of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable lightchain sequence that is at least about 80%, 85%, 90%,
-108-95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs 171, 179, 187, 195, 203, 208 or 210.
In another embodiment, the invention is directed to an isolated polynucleotide encoding an antibody binding to DR5 of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable heavy chain sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs 167, 175, 183, 191 or 199.
In another embodiment, the invention is directed to an isolated polynucleotide encoding an antibody binding to DRS of the invention or a fragment thereof, wherein the polynucleotide comprises a sequence that encodes a variable light chain sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence in SEQ ID NOs 171, 179, 187, 195 or 203.
In certain embodiments the polynucleotide or nucleic acid is DNA. In other embodiments, a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA
(mRNA). RNA of the present invention may be single stranded or double stranded.
In further objects the present invention relates to an expression vector comprising a nucleic acid sequence of the present invention and to a prokaryotic or eukaryotic host cell comprising a vector of the present invention. In addition a method of producing an antibody comprising culturing the host cell so that the antibody is produced is provided.
G. Antibody Variants In certain embodiments, amino acid sequence variants of the bispecific antibodies and antibodies binding to DRS provided herein are contemplated, in addition to those described above. For example, it may be desirable to improve the binding affinity and/or other biological properties of the bispecific antibody or the antibody binding to DRS. Amino acid sequence variants of a bispecific antibody or an antibody binding to DRS may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the bispecific antibody or the antibody binding to DRS, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
-109-1. Substitution, Insertion, and Deletion Variants In certain embodiments, antibody variants having one or more amino acid substitutions are provided. Sites of interest for substitutional mutagenesis include the HVRs and FRs.
Conservative substitutions are shown in Table B under the heading of "conservative substitutions." More substantial changes are provided in Table B under the heading of "exemplary substitutions," and as further described below in reference to amino acid side chain classes. Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved ADCC or CDC.
TABLE B
Original Exemplary Preferred Residue Substitutions Substitutions Ala (A) Val; Leu; Ile Val Arg (R) Lys; Gln; Asn Lys Asn (N) Gln; His; Asp, Lys; Arg Gln Asp (D) Glu; Asn Glu Cys (C) Ser; Ala Ser Gln (Q) Asn; Glu Asn Glu (E) Asp; Gln Asp Gly (G) Ala Ala His (H) Asn; Gln; Lys; Arg Arg He (I) Leu; Val; Met; Ala; Phe; Norleucine Leu Leu (L) Norleucine; Ile; Val; Met; Ala; Phe Ile Lys (K) Arg; Gln; Asn Arg Met (M) Leu; Phe; Ile Leu Phe (F) Trp; Leu; Val; Ile; Ala; Tyr Tyr Pro (P) Ala Ala Ser (S) Thr Thr Thr (T) Val; Ser Ser Trp (W) Tyr; Phe Tyr Tyr (Y) Trp; Phe; Thr; Ser Phe Val (V) Ile; Leu; Met; Phe; Ala; Norleucine Leu Amino acids may be grouped according to common side-chain properties:
-110-(1) hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile;
(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gln;
(3) acidic: Asp, Glu;
(4) basic: His, Lys, Arg;
(5) residues that influence chain orientation: Gly, Pro;
(6) aromatic: Trp, Tyr, Phe.
Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
One type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody). Generally, the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody. An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
Alterations (e.g., substitutions) may be made in HVRs, e.g., to improve antibody affinity.
Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH
or VL being tested for binding affinity. Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom et al. in Methods in Molecular Biology 178:1-37 (O'Brien et al., ed., Human Press, Totowa, NJ, (2001).) In some embodiments of affinity maturation, diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis). A secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity. Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
-111-In certain embodiments, substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen. For example, conservative alterations (e.g., conservative substitutions as provided herein) that do not substantially reduce binding affinity may be made in HVRs.
Such alterations may be outside of HVR "hotspots" or SDRs. In certain embodiments of the variant VH and VL
sequences provided above, each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
A useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells (1989) Science, 244:1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced by a neutral or negatively charged amino acid (e.g., alanine or polyalanine) to determine whether the interaction of the antibody with antigen is affected. Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions.
Alternatively, or additionally, a crystal structure of an antigen-antibody complex to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.
Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue. Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
2. Glycosylation variants In certain embodiments, a bispecific antibody or an antibody binding to DR5 provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
Where the bispecific antibody or the antibody binding to DRS comprises an Fc region, the carbohydrate attached thereto may be altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH
-112-15:26-32 (1997). The oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (G1cNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure. In some embodiments, modifications of the oligosaccharide in a bispecific antibody or an antibody binding to DR5 of the invention may be made in order to create antibody variants with certain improved properties.
In one embodiment, bispecific antibody variants or variants of antibodies binding to DRS
are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%. The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC
function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.);

(Kyowa Hakko Kogyo Co., Ltd). Examples of publications related to "defucosylated" or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739;
WO
2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US
2004/0132140; US
2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO
2003/084570;
WO 2005/035586; WO 2005/035778; W02005/053742; W02002/031140; Okazaki et al.
J. MoL
Biol. 336:1239-1249 (2004); Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004). Examples of cell lines capable of producing defucosylated antibodies include Lec13 CHO
cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986);
US Pat Appl No US 2003/0157108 Al, Presta, L; and WO 2004/056312 Al, Adams et al., especially at Example 11), and knockout cell lines, such as alpha-1,6-fucosyltransferase gene, FUT8, knockout CHO
cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al., Biotechnol. Bioeng., 94(4):680-688 (2006); and W02003/085107).
Bispecific antibodies variants or variants of antibodies binding to DRS are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the bispecific antibody or the antibody binding to DRS is bisected by GlcNAc.
Such bispecific antibody variants or variants of antibodies binding to DRS may have reduced
-113-fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean-Mairet et al.); US Patent No.
6,602,684 (Umana et al.); and US 2005/0123546 (Umana et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO

(Patel et al.); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
3. Cysteine engineered antibody variants In certain embodiments, it may be desirable to create cysteine engineered bispecific antibodies or antibodies binding to DR5, e.g., "thioMAbs," in which one or more residues of a bispecific antibody or antibodies binding to DR5 are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the bispecific antibody or the antibody binding to DR5. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate. In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; A118 (EU
numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No. 7,521,541.
H. Recombinant Methods and Compositions Bispecific antibodies and antibodies binding to DRS of the invention may be obtained, for example, by solid-state peptide synthesis (e.g. Merrifield solid phase synthesis) or recombinant production. For recombinant production one or more polynucleotide encoding the bispecific antibodies or antibodies binding to DRS (or fragments), e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such polynucleotide may be readily isolated and sequenced using conventional procedures. In one embodiment a vector, preferably an expression vector, comprising one or more of the polynucleotides of the invention is provided. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the coding sequence of a bispecific antibody (fragment) or an antibody (fragment) binding to DRS along with appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA
techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al., MOLECULAR CLONING: A
LABORATORY
-114-MANUAL, Cold Spring Harbor Laboratory, N.Y. (1989); and Ausubel et al., CURRENT
PROTOCOLS IN MOLECULAR BIOLOGY, Greene Publishing Associates and Wiley Interscience, N.Y (1989). The expression vector can be part of a plasmid, virus, or may be a nucleic acid fragment. The expression vector includes an expression cassette into which the polynucleotide encoding the bispecific antibody (fragment) or an antibody (fragment) binding to DR5 (i.e. the coding region) is cloned in operable association with a promoter and/or other transcription or translation control elements. As used herein, a "coding region" is a portion of nucleic acid which consists of codons translated into amino acids. Although a "stop codon" (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, if present, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, 5 and 3' untranslated regions, and the like, are not part of a coding region.
Two or more coding regions can be present in a single polynucleotide construct, e.g. on a single vector, or in separate polynucleotide constructs, e.g. on separate (different) vectors. Furthermore, any vector may contain a single coding region, or may comprise two or more coding regions, e.g.
a vector of the present invention may encode one or more polypeptides, which are post- or co-translationally separated into the final proteins via proteolytic cleavage. In addition, a vector, polynucleotide, or nucleic acid of the invention may encode heterologous coding regions, either fused or unfused to a polynucleotide encoding the bispecific antibody (fragment) or an antibody (fragment) binding to DRS of the invention, or variant or derivative thereof.
Heterologous coding regions include without limitation specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain. An operable association is when a coding region for a gene product, e.g. a polypeptide, is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s). Two DNA fragments (such as a polypeptide coding region and a promoter associated therewith) are "operably associated" if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not interfere with the ability of the expression regulatory sequences to direct the expression of the gene product or interfere with the ability of the DNA template to be transcribed. Thus, a promoter region would be operably associated with a nucleic acid encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid. The promoter may be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells. Other transcription control elements, besides a promoter, for example enhancers, operators, repressors, and transcription termination
-115-signals, can be operably associated with the polynucleotide to direct cell-specific transcription.
Suitable promoters and other transcription control regions are disclosed herein. A variety of transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions, which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (e.g. the immediate early promoter, in conjunction with intron-A), simian virus 40 (e.g. the early promoter), and retroviruses (such as, e.g. Rous sarcoma virus). Other transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit a-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as inducible promoters (e.g. promoters inducible tetracyclins). Similarly, a variety of translation control elements are known to those of ordinary skill in the art. These include, but are not limited to ribosome binding sites, translation initiation and termination codons, and elements derived from viral systems (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence). The expression cassette may also include other features such as an origin of replication, and/or chromosome integration elements such as retroviral long terminal repeats (LTRs), or adeno-associated viral (AAV) inverted terminal repeats (ITRs).
Polynucleotide and nucleic acid coding regions of the present invention may be associated with additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide of the present invention. For example, if secretion of the bispecific antibody or the antibody binding to DR5 is desired, DNA
encoding a signal sequence may be placed upstream of the nucleic acid encoding a bispecific antibody of the invention or the antibody binding to DRS of the invention or a fragment thereof. According to the signal hypothesis, proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated. Those of ordinary skill in the art are aware that polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the translated polypeptide to produce a secreted or "mature" form of the polypeptide. In certain embodiments, the native signal peptide, e.g. an immunoglobulin heavy chain or light chain signal peptide is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it. Alternatively, a heterologous mammalian signal
-116-peptide, or a functional derivative thereof, may be used. For example, the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator (TPA) or mouse P-glucuronidase.
DNA encoding a short protein sequence that could be used to facilitate later purification (e.g. a histidine tag) or assist in labeling the bispecific antibody or the antibody binding to DR5 may be included within or at the ends of the bispecific antibody (fragment) or the antibody (fragment) binding to DRS encoding polynucleotide.
In a further embodiment, a host cell comprising one or more polynucleotides of the invention is provided. In certain embodiments a host cell comprising one or more vectors of the invention is provided. The polynucleotides and vectors may incorporate any of the features, singly or in combination, described herein in relation to polynucleotides and vectors, respectively. In one such embodiment a host cell comprises (e.g. has been transformed or transfected with) a vector comprising a polynucleotide that encodes (part of) a bispecific antibody or an antibody binding to DRS of the invention. As used herein, the term "host cell" refers to any kind of cellular system which can be engineered to generate the bispecific antibodies or an antibody binding to DRS of the invention or fragments thereof. Host cells suitable for replicating and for supporting expression of bispecific antibodies or of antibodies binding to DRS are well known in the art.
Such cells may be transfected or transduced as appropriate with the particular expression vector and large quantities of vector containing cells can be grown for seeding large scale fermenters to obtain sufficient quantities of the bispecific antibody or of the antibodies binding to DRS for clinical applications. Suitable host cells include prokaryotic microorganisms, such as E. coli, or various eukaryotic cells, such as Chinese hamster ovary cells (CHO), insect cells, or the like. For example, polypeptides may be produced in bacteria in particular when glycosylation is not needed. After expression, the polypeptide may be isolated from the bacterial cell paste in a soluble fraction and can be further purified. In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized", resulting in the production of a polypeptide with a partially or fully human glycosylation pattern.
See Gerngross, Nat Biotech 22, 1409-1414 (2004), and Li et al., Nat Biotech 24, 210-215 (2006).
Suitable host cells for the expression of (glycosylated) polypeptides are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells. Plant
-117-cell cultures can also be utilized as hosts. See e.g. US Patent Nos.
5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTm technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts.
For example, mammalian cell lines that are adapted to grow in suspension may be useful.
Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7);
human embryonic kidney line (293 or 293T cells as described, e.g., in Graham et al., J Gen Virol 36, 59 (1977)), baby hamster kidney cells (BHK), mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol Reprod 23, 243-251 (1980)), monkey kidney cells (CV1), African green monkey kidney cells (VERO-76), human cervical carcinoma cells (HELA), canine kidney cells (MDCK), buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver cells (Hep G2), mouse mammary tumor cells (MMT 060562), TRI cells (as described, e.g., in Mather et al., Annals N.Y. Acad Sci 383, 44-68 (1982)), MRC 5 cells, and F54 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including dhfr- CHO
cells (Urlaub et al., Proc Natl Acad Sci USA 77, 4216 (1980)); and myeloma cell lines such as YO, NSO, P3X63 and 5p2/0. For a review of certain mammalian host cell lines suitable for protein production, see, e.g., Yazaki and Wu, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ), pp. 255-268 (2003). Host cells include cultured cells, e.g., mammalian cultured cells, yeast cells, insect cells, bacterial cells and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue. In one embodiment, the host cell is a eukaryotic cell, preferably a mammalian cell, such as a Chinese Hamster Ovary (CHO) cell, a human embryonic kidney (HEK) cell or a lymphoid cell (e.g., YO, NSO, 5p20 cell).
Standard technologies are known in the art to express foreign genes in these systems. Cells expressing a polypeptide comprising either the heavy or the light chain of an antigen binding domain such as an antibody, may be engineered so as to also express the other of the antibody chains such that the expressed product is an antibody that has both a heavy and a light chain.
In one embodiment, a method of producing a bispecific antibody or an antibody binding to DR5 according to the invention is provided, wherein the method comprises culturing a host cell comprising a polynucleotide encoding the bispecific antibody or the antibody binding to DR5, as provided herein, under conditions suitable for expression of the bispecific antibody or the antibody binding to DR5, and recovering the bispecific antibody or the antibody binding to DRS
from the host cell (or host cell culture medium).
-118-The components of the bispecific antibody or the antibody binding to DR5 are genetically fused to each other. Bispecific antibodies or the antibodies binding to DRS can be designed such that its components are fused directly to each other or indirectly through a linker sequence. The composition and length of the linker may be determined in accordance with methods well known in the art and may be tested for efficacy. Examples of linker sequences between different components of bispecific antibodies are found in the sequences provided herein. Additional sequences may also be included to incorporate a cleavage site to separate the individual components of the fusion if desired, for example an endopeptidase recognition sequence.
In certain embodiments the Fab fragments forming part of the bispecific antibody or the antibody binding to DRS comprise at least an antibody variable region capable of binding an antigenic determinant. Variable regions can form part of and be derived from naturally or non-naturally occurring antibodies and fragments thereof. Methods to produce polyclonal antibodies and monoclonal antibodies are well known in the art (see e.g. Harlow and Lane, "Antibodies, a laboratory manual", Cold Spring Harbor Laboratory, 1988). Non-naturally occurring antibodies can be constructed using solid phase-peptide synthesis, can be produced recombinantly (e.g. as described in U.S. patent No. 4,186,567) or can be obtained, for example, by screening combinatorial libraries comprising variable heavy chains and variable light chains (see e.g. U.S.
Patent. No. 5,969,108 to McCafferty).
Any animal species of antibody, antibody fragment, antigen binding domain or variable region can be used in the bispecific antibodies or the antibodies binding to DRS of the invention. Non-limiting antibodies, antibody fragments, antigen binding domains or variable regions useful in the present invention can be of murine, primate, or human origin. If the bispecific antibody or the antibody binding to DRS is intended for human use, a chimeric form of antibody may be used wherein the constant regions of the antibody are from a human. A humanized or fully human form of the antibody can also be prepared in accordance with methods well known in the art (see e. g. U.S. Patent No. 5,565,332 to Winter). Humanization may be achieved by various methods including, but not limited to (a) grafting the non-human (e.g., donor antibody) CDRs onto human (e.g. recipient antibody) framework and constant regions with or without retention of critical framework residues (e.g. those that are important for retaining good antigen binding affinity or antibody functions), (b) grafting only the non-human specificity-determining regions (SDRs or a-CDRs; the residues critical for the antibody-antigen interaction) onto human framework and constant regions, or (c) transplanting the entire non-human variable domains, but "cloaking"
them with a human-like section by replacement of surface residues. Humanized antibodies and
-119-methods of making them are reviewed, e.g., in Almagro and Fransson, Front Biosci 13, 1619-1633 (2008), and are further described, e.g., in Riechmann et al., Nature 332, 323-329 (1988);
Queen et al., Proc Natl Acad Sci USA 86, 10029-10033 (1989); US Patent Nos.
5,821,337, 7,527,791, 6,982,321, and 7,087,409; Jones et al., Nature 321, 522-525 (1986);
Morrison et al., Proc Natl Acad Sci 81, 6851-6855 (1984); Morrison and 0i, Adv Immunol 44, 65-92 (1988);
Verhoeyen et al., Science 239, 1534-1536 (1988); Padlan, Molec Immun 31(3), 169-217 (1994);
Kashmiri et al., Methods 36, 25-34 (2005) (describing SDR (a-CDR) grafting);
Padlan, Mol Immunol 28, 489-498 (1991) (describing "resurfacing"); Dall'Acqua et al., Methods 36, 43-60 (2005) (describing "FR shuffling"); and Osbourn et al., Methods 36, 61-68 (2005) and Klimka et al., Br J Cancer 83, 252-260 (2000) (describing the "guided selection"
approach to FR shuffling).
Human antibodies and human variable regions can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr Opin Pharmacol 5, 368-74 (2001) and Lonberg, Curr Opin Immunol 20, 450-459 (2008).
Human variable regions can form part of and be derived from human monoclonal antibodies made by the hybridoma method (see e.g. Monoclonal Antibody Production Techniques and Applications, pp.
51-63 (Marcel Dekker, Inc., New York, 1987)). Human antibodies and human variable regions may also be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge (see e.g. Lonberg, Nat Biotech 23, 1117-1125 (2005). Human antibodies and human variable regions may also be generated by isolating Fv clone variable region sequences selected from human-derived phage display libraries (see e.g., Hoogenboom et al. in Methods in Molecular Biology 178, 1-37 (O'Brien et al., ed., Human Press, Totowa, NJ, 2001); and McCafferty et al., Nature 348, 552-554; Clackson et al., Nature 352, 624-628 (1991)).
Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
In certain embodiments, the Fab fragments useful in the present invention are engineered to have enhanced binding affinity according to, for example, the methods disclosed in U.S. Pat. Appl.
Publ. No. 2004/0132066, the entire contents of which are hereby incorporated by reference. The ability of the bispecific antibody or the antibody binding to DR5 of the invention to bind to a specific antigenic determinant can be measured either through an enzyme-linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g.
surface plasmon resonance technique (analyzed on a BIACORE T100 system) (Liljeblad, et al., Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)).
-120-Competition assays may be used to identify an antibody, antibody fragment, antigen binding domain or variable domain that competes with a reference antibody for binding to a particular antigen. In certain embodiments, such a competing antibody binds to the same epitope (e.g. a linear or a conformational epitope) that is bound by the reference antibody.
Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ). In an exemplary competition assay, immobilized antigen is incubated in a solution comprising a first labeled antibody that binds to the antigen and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to the antigen. The second antibody may be present in a hybridoma supernatant. As a control, immobilized antigen is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to the antigen, excess unbound antibody is removed, and the amount of label associated with immobilized antigen is measured. If the amount of label associated with immobilized antigen is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to the antigen. See Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY).
Bispecific antibodies or antibodies binding to DR5 prepared as described herein may be purified by art-known techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography, and the like. The actual conditions used to purify a particular protein will depend, in part, on factors such as net charge, hydrophobicity, hydrophilicity etc., and will be apparent to those having skill in the art. For affinity chromatography purification an antibody, ligand, receptor or antigen can be used to which the bispecific antibody or the antibody binding to DRS binds. For example, for affinity chromatography purification of bispecific antibodies of the invention, a matrix with protein A or protein G may be used. Sequential Protein A or G
affinity chromatography and size exclusion chromatography can be used to isolate a bispecific antibody essentially as described in the Examples. The purity of the bispecific antibody or the antibody binding to DRS can be determined by any of a variety of well known analytical methods including gel electrophoresis, high pressure liquid chromatography, and the like.
-121-I. Assays Bispecific antibodies that bind to DRS and FAP and antibodies binding to DRS
provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art 1. Affinity assays The affinity of the bispecific antibody and the antibody binding to DRS
provided therein for DRS
and/ or FAP can be determined in accordance with the methods set forth in the Examples by surface plasmon resonance (SPR), using standard instrumentation such as a BIAcore instrument (GE Healthcare), and receptors or target proteins such as may be obtained by recombinant expression. Alternatively, binding of bispecific antibody and the antibody binding to DRS
provided therein to DRS and/or FAP may be evaluated using cell lines expressing the particular receptor or target antigen, for example by flow cytometry (FACS). A specific illustrative and exemplary embodiment for measuring binding affinity is described in the following and in the Examples below.
According to one embodiment, KD is measured by surface plasmon resonance using a BIACORE T100 machine (GE Healthcare) at 25 C.
To analyze the interaction between the Fc-portion and Fc receptors, His-tagged recombinant Fc-receptor is captured by an anti-Penta His antibody (Qiagen) immobilized on CMS
chips and the bispecific constructs are used as analytes. Briefly, carboxymethylated dextran biosensor chips (CMS, GE Healthcare) are activated with N-ethyl-N'-(3-dimethylaminopropy1)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions.
Anti Penta-His antibody is diluted with 10 mM sodium acetate, pH 5.0, to 40 ug/m1 before injection at a flow rate of 5 .t1/min to achieve approximately 6500 response units (RU) of coupled protein. Following the injection of the ligand, 1 M ethanolamine is injected to block unreacted groups. Subsequently the Fc-receptor is captured for 60 s at 4 or 10 nM. For kinetic measurements, four-fold serial dilutions of the bispecific construct (range between 500 nM and 4000 nM) are injected in HBS-EP (GE Healthcare, 10 mM HEPES, 150 mM NaC1, 3 mM
EDTA, 0.05 % Surfactant P20, pH 7.4) at 25 C at a flow rate of 30 .i1/min for 120 s.
To determine the affinity to the target antigen, bispecific constructs are captured by an anti human Fab specific antibody (GE Healthcare) that is immobilized on an activated CMS-sensor chip surface as described for the anti Penta-His antibody. The final amount of coupled protein is is approximately 12000 RU. The bispecific constructs are captured for 90 s at 300 nM. The
-122-target antigens are passed through the flow cells for 180 s at a concentration range from 250 to 1000 nM with a flowrate of 30 .i1/min. The dissociation is monitored for 180 s.
Bulk refractive index differences are corrected for by subtracting the response obtained on reference flow cell. The steady state response was used to derive the dissociation constant KD by non-linear curve fitting of the Langmuir binding isotherm. Association rates (kor,) and dissociation rates (kc,ff) are calculated using a simple one-to-one Langmuir binding model (BIACORE T100 Evaluation Software version 1.1.1) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) is calculated as the ratio koff/kon. See, e.g., Chen et al., J Mol Biol 293, 865-881 (1999).
2. Binding assays and other assays In one aspect, a bispecific antibody or an antibody that binds to DR5 of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, Western blot, etc.
In another aspect, competition assays may be used to identify an antibody that competes with a specific anti-FAP antibody or a specific anti-DR5 antibody for binding to FAP or DR5 respectively. In certain embodiments, such a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by a specific anti-FAP
antibody or a specific anti-DR5 antibody. Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ). Further methods are described in the example section.
3. Activity assays In one aspect, assays are provided for identifying bispecific antibodies that bind to DRS
and FAP or antibodies that binds to DRS thereof having biological activity.
Biological activity may include, e.g., DNA fragmentation, induction of apoptosis and lysis of targeted cells.
Antibodies having such biological activity in vivo and/or in vitro are also provided.
In certain embodiments, a bispecific antibody or an antibodiy that binds to DRS of the invention is tested for such biological activity. Assays for detecting cell lysis (e.g. by measurement of LDH release) or apoptosis (e.g. using the TUNEL assay) are well known in the art. Assays for measuring ADCC or CDC are also described in WO 2004/065540 (see Example 1 therein), the entire content of which is incorporated herein by reference.
-123-J. Pharmaceutical Formulations Pharmaceutical formulations of a bispecific antibody that binds to DR5 and FAP
or an antibody that binds to DR5 as described herein are prepared by mixing such bispecific antibody or antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride;
phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol;
resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins;
hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include insterstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rHuPH20 (HYLENEX , Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
Exemplary lyophilized antibody formulations are described in US Patent No.
6,267,958.
Aqueous antibody formulations include those described in US Patent No.
6,171,586 and W02006/044908, the latter formulations including a histidine-acetate buffer.
The formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
-124-Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
The formulations to be used for in vivo administration are generally sterile.
Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
K. Therapeutic Methods and Compositions Any of the bispecific antibodies that bind to DR5 and FAP and the novel antibodies binding to DR5 provided herein may be used in therapeutic methods.
In one aspect, a bispecific antibody that binds to DR5 and FAP for use as a medicament is provided. In further aspects, a bispecific antibody that binds to DR5 and FAP use in treating cancer is provided. In certain embodiments, a bispecific antibody that binds to DR5 and FAP for use in a method of treatment is provided. In certain embodiments, the invention provides a bispecific antibody that binds to DRS and FAP for use in a method of treating an individual having cancer comprising administering to the individual an effective amount of the bispecific antibody that binds to DRS and FAP. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. An "individual" according to any of the above embodiments is preferably a human. In one preferred embodiment said cancer is pancreatic cancer or colorectal carcinoma.
In one aspect, an antibody that binds to DRS for use as a medicament is provided. In further aspects, a antibody that binds to DRS use in treating cancer is provided. In certain embodiments, a antibody that binds to DRS for use in a method of treatment is provided. In certain embodiments, the invention provides an antibody that binds to DRS for use in a method of treating an individual having cancer comprising administering to the individual an effective amount of the antibody that binds to DRS. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. An "individual" according to any of the above
-125-embodiments is preferably a human. In one preferred embodiment said cancer is pancreatic cancer or colorectal carcinoma.
In a further aspect, the invention provides for the use of a bispecific antibody that binds to DR5 and FAP in the manufacture or preparation of a medicament. In another aspect, the invention provides for the use of an antibody that binds to DRS in the manufacture or preparation of a medicament. In one embodiment, the medicament is for treatment of cancer.
In a further embodiment, the medicament is for use in a method of treating cancer comprising administering to an individual having cancer an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. An "individual"
according to any of the above embodiments may be a human.
In a further aspect, the invention provides a method for treating cancer. In one embodiment, the method comprises administering to an individual having cancer an effective amount of a bispecific antibody that binds to DRS and FAP or of a novel antibody binding to DRS. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below. An "individual"
according to any of the above embodiments may be a human. In one preferred embodiment said cancer is pancreatic cancer or colorectal carcinoma.
In a further aspect, the invention provides pharmaceutical formulations comprising any of the bispecific antibodies that bind to DRS and FAP provided herein, e.g., for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the bispecific antibodies that bind to DRS and FAP provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the bispecific antibodies that bind to DRS and FAP provided herein and at least one additional therapeutic agent, e.g., as described below.
In a further aspect, the invention provides pharmaceutical formulations comprising any of the antibodies that bind to DRS provided herein, e.g., for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the antibodies that bind to DRS provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the antibodies that bind to DRS
provided herein and at least one additional therapeutic agent, e.g., as described below.
-126-A bispecific antibody or a novel antibody binding to DR5 of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
Bispecific antibodies or novel antibodies binding to DRS of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The bispecific antibody or the novel antibody binding to DRS need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
For the prevention or treatment of disease, the appropriate dosage of a bispecific antibody or a novel antibody binding to DRS of the invention will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the bispecific antibody or the novel antibody binding to DRS is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the bispecific antibody or to the novel antibody binding to DRS, and the discretion of the attending physician. The bispecific antibody or the novel antibody binding to DRS is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 p g/kg to 15 mg/kg (e.g. 0.1mg/kg-10mg/kg) of the bispecific antibody or the novel antibody binding to DRS can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 p g/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on
-127-the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the bispecific antibody or the novel antibody binding to DR5 would be in the range from about 0.05 mg/kg to about 10 mg/kg.
Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g.
about six doses of the bispecific antibody or of the novel antibody binding to DRS). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
It is understood that any of the above formulations or therapeutic methods may be carried out using an immunoconjugate of the invention in place of or in addition to a bispecific antibody that binds to DR5 and FAP or a novel antibody binding to DR5 of the invention.
L. Articles of Manufacture In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a bispecific antibody or a novel antibody binding to DRS of the invention. The label or package insert indicates that the composition is used for treating the condition of choice.
Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a bispecific antibody or a novel antibody binding to DRS of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection
-128-(BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
It is understood that any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to a bispecific antibody that binds to DR5 and FAP or a novel antibody binding to DRS of the invention.
III. EXAMPLES
The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.
Example 1: A DR5 ¨ FAP death receptor agonistic bispecific antibody is able to mediate apoptosis of one cell line via cross-linking by a second cell line.
One approach of induction of apoptosis by cross-linking of death receptors as DRS (apart from cross-linking via an antigen expressed by the tumor cell), is targeting the stroma surrounding the tumor. In that case the targeted antigen is not displayed directly by the tumor cells but by a second, different cell type. One example for this kind of antigen would be FAP
(fibroblast activation protein). This protein is expressed on activated fibroblast as they are found in the tumor stroma.
To investigate the possibilities of tumor targeted induction of apoptosis using bispecific death receptor agonistic antibodies targeting human DRS and an antigen from the tumor stroma, bispecific molecules were generated that consist of an IgG1 part that recognizes DRS and a FAP
binding scFv that is fused to the C-terminus of the heavy chain of the antibody. The sequence of the DRS targeting IgG was taken from the Drozitumab sequence as described in 0031414 Al. The sequence of variable heavy and light chain of the FAP binding scFv moieties were taken from Fab anti FAP molecules (3F2 or 4G8) isolated by phage display as described in W02012/020006A1. The FAP scFvs are fused by a (G4S)2 connector to the C-terminus of the anti DRS IgG heavy chain or light chain. Both anti FAP antibodies bind to different epitopes on
-129-fibroblast activation protein (FAP) and are cross-reactive with the human, murine and cynomolgus antigens.
Table 1: Description of tested bispecific DR5-FAP molecules with C-terminal fusion of FAP binding scFv's to DRS heavy or light chain. Linker and connector length and purification yields are given.
Molecule I )escription Connector Linker Yield in sclv Inig/1.1 3F2-scFv_HC Fusion of disulfide stabilized (G4S)2 (G4S)4 2.53 scFv (H44L100) to C-terminus (SEQ ID NO.:111) of Drozitumab heavy chain 4G8-scFv_HC Fusion of disulfide stabilized (G45)2 (G45)4 4.29 scFv (H44L100) to C-terminus (SEQ ID NO.:112) of Drozitumab heavy chain 4G8-scFv_LC Fusion of disulfide stabilized (G45)2 (G45)4 2.15 scFv (H44L100) to C-terminus (SEQ ID NO.:113) of Drozitumab light chain In this kind of setting two different cell lines have to be used for the in vitro activity assays: one cell line (the target cell line) should express human DR5, has to be apoptosis competent but does not need to express FAP. The second cell line (the effector cell line) has to be apoptosis negative (either by apoptosis resistance or by not expressing DR5) but needs to express FAP on the surface.
One possible effector cell line that fulfills the desired criteria is the human fibroblast cell line GM05389. As shown in figure la this cell line expresses significant levels of FAP compared to the cell line 5W872 which only showed FAP expression with the highest tested antibody concentration (10 pg / ml) but does not undergo apoptosis by non-cross-linked Drozitumab as seen in figure lb. Therefore this cell line seems to be a potential effector cell line in an apoptosis assay where DNA fragmentation of a target cell line is induced by cross-linking via an antigen expressed on a second cell line.
As a target cell line the human kidney-adenocarcinoma cell line ACHN or the human breast cancer cell line MDA-MB-231 were used. They express comparable, low levels of DRS
(Figure 2) and are sensitive to DRS mediated apoptosis induction. In figure 3 the results of
-130-induction of DNA fragmentation of ACHN cells compared to the combination of ACHN and GM05389 cell lines by tumor targeted cross-linking of DRS via FAP is summarized. The control antibody (Drozitumab) induces apoptosis upon cross-linking via a secondary antibody that targets the Fc part (white bars) in both cell lines, but shows also some activity without crosslinking. The bispecific DRS-FAP molecules show significant induction of apoptosis in the presence of both ¨ target and effector cell line (black bars). In the absence of FAP expressing fibroblasts (GM05389) there is a slightly increased apoptosis compared to non-cross-linked Drozitumab (grey bars). We interpret this result in a way that the DRS
receptors on ACHN cells are cross-linked upon binding to the FAP antigen expressed by the fibroblast cell line GM05389.
All molecules show comparable maximal apoptosis activity.
Example 2: DR5 bispecific agonistic antibodies with cross-reactive FAP binders in scFab format fused to Drozitumab demonstrate apoptosis activity in co-culture assays As demonstrated in example 1 DRS-FAP bispecific agonistic antibodies in which the FAP
targeting moiety in scFv format is fused to the C-terminus of the Drozitumab heavy or light chain (2x2 format) are able to induce apoptosis in a two cell line co-culture setting in which one cell line expresses FAP (effector cells) while the second cell line serves as the DRS receptor target.
Since these molecules containing an scFv fusion showed some disadvantageous properties (low production yield and tendency to form aggregates), constructs were generated in which the FAP binding unit was replaced by single chain Fab entities (scFab), fused to the C-terminus of either the heavy or light chain of Drozitumab leading to the production of four different molecules as described in table 2. The connection of the scFab units to the IgG part of the bispecific molecules occurs via a (G4S)4 sequence whereas the scFab internal linker consists of 32 amino acids.
These molecules were transiently produced by standard recombinant technologies and purified in sufficient amounts and good quality for detailed testing of FAP
and DRS binding (FACS; Biacore, not shown).
Table 2: Description and characterization of the different Drozitumab-FAP
(scFab) fusions.
In all molecules the anti FAP scFab is fused by a 20 mer connector ((G45)4) to Drozitumab.
Name Description Yield Connector [mg / L]
-131-GA803_AOl_B 01A_004 scFab: 3F2; VLCL-VHCH1 3.9 (G4S)4 3F2_scFab_HC C-terminal fusion to Drozitumab heavy chain (2x2) (SEQ ID NO.:114) GA803_AO 1 _B 01B_005 scFab: 3F2; VLCL-VHCH1 4.9 (G4S)4 3F2_scFab_LC C-terminal fusion to Drozitumab light chain (2x2) (SEQ ID NO.:115) GA803_AO1_BO2A_001 scFab: 4G8; VLCL-VHCH1 2.0 (G4S)4 4G8_scFab_HC C-terminal fusion to Drozitumab heavy chain (2x2) (SEQ ID NO.:116) GA803_AO1_BO2B_002 scFab: 4G8; VLCL-VHCH1 2.5 (G45)4 4G8_scFab_LC C-terminal fusion to Drozitumab light chain (2x2) (SEQ ID NO.:117) Analysis of apoptosis induction of a DR5 expressing target cell line (e.g. the renal carcinoma cell line ACHN) in the presence and absence of a second, FAP
expressing 'effector' cell line (e.g. fibroblast line GM05389) is shown in figure 4a and 4b.The apoptosis induction was analysed with a DNA fragmentation assay. In the assays a ratio of target to effector cells of 1 : 1 was used and DNA fragmentation after 24 hrs of co-culture in the presence of Drozitumab, hyper-cross-linked Drozitumab or the bispecific constructs (all used in concentrations of 1 tg /
ml and 0.1 pg / ml) was analyzed.
For all tested constructs it could be demonstrated that the bispecific molecules, independent of the used FAP binder (either 3F2 or 4G8) as scFabs fused to Drozitumab show increased apoptosis induction in ACHN target cells only in the presence of the fibroblast effector cell line. However, at high concentrations of the bispecific molcules a low degree of apoptosis activity could be observed in the absence of FAP expressing fibroblasts. In general and independent on the used FAP binder, molecules with scFab fusion to the C-terminus of the heavy chain were more active as molecules where the scFabs were fused to the C-terminus of the light chain. In addition, 4G8 containing constructs seemed to be more potent than 3F2 containing
-132-molecules. However, activity of scFab based formats was lower compared to the analogous scFv based constructs.
Drozitumab without additional cross-linking by anti Fc antibodies showed a low apoptosis induction. However, Drozitumab hyper-cross-linked via a secondary anti Fc antibody alone - a highly artificial situation - revealed the highest apoptosis activity which could not been reached by any of the tested molecules.
Example 3: DR5 - FAP bispecific agonistic antibodies in CrossFab format demonstrate superior characteristics over scFab containing molecules Since the evaluated scFab containing bispecific molecules still showed some disadvantageous characteristics (e.g. low expression yield, optimizable apoptosis activity) a novel format that should overcome these liabilities was analyzed: fusion of a FAP binder (4G8) in CrossFab format to the C-terminus of Drozitumab heavy chain. In this format the FAP binding moiety is used in a 'crossover' exchange of variable regions in the Fab fragment as described in table 3 and shown in figure 5. The CrossFab molecules are linked via a 20mer connector ((G4S)4) to the Drozitumab heavy chain.
Table 3: Description and production yields of bispecific Drozitumab ¨ FAP
CrossFab constructs in two different formats.
Figure Name Description Yield [mg /
Monomer [%[
5a GA803_A01_E02A_014 = Fusion of 4G8 VH-CL to 36.1 C-terminus of Drozitumab Drozitumab¨X ¨ FAP_A
heavy chain (SEQ
(2+2) ID NO.:118) 100.0 (SEQ ID NO.:118, = Separate VL-CH1 cassette 119,120) (SEQ ID NO.:120) = Separate Drozitumab light chain cassette (SEQ
ID NO.:119) 5b GA803_A01_E02A_015 = Fusion of 4G8 VL-CH1 to 16.7 C-terminus of Drozitumab
-133-Drozitumab¨X ¨ FAP_B heavy chain (SEQ ID
(2+2) NO.:121) 100.0 (SEQ ID NO.:121, 122, = Separate VH-CL cassette 119) (SEQ ID NO.:122) = Separate Drozitumab light chain cassette (SEQ
ID NO.:119) Both CrossFab molecules (also see figure 5 for organization of the constructs) were transiently produced in HEK293 EBNA cells (either in adherent or suspension cells) and purified with standard methods. Compared to scFab and scFv containing bispecific constructs the CrossFab molecules exhibited significantly increased expression levels leading to approximately 10-fold higher product yields with acceptable low aggregate contents as shown in table 4.
Figure 6 demonstrates the aggregate content during production of different bispecific DR5-FAP molecules. Chromatograms from preparative size exclusion chromatography during purification are shown for the scFv (A), scFab (B) and CrossFab (C) fusion to the C-terminus of Drozitumab heavy chain. The scFv and scFab fusion constructs show significantly higher aggregate contents compared to the CrossFab containing molecule. Removal of these aggregates during purification leads to huge loss of material which can easily be seen from the yields obtained after purification.
Binding of the molecules to human FAP, expressed on recombinant HEK293 cells was detected by FACS analysis. Figure 7 shows the FACS binding results of two Drozitumab-CrossFab molecules (Drozitumab¨X¨FAP_A, dotted bar and Drozitumab¨X¨FAP_B, white bar) compared to an analogous scFab construct (hatched bar) or the corresponding FAP (4G8) IgG
molecule (black bar). All three bispecific molecules in which the 4G8 FAP
binding moiety is fused to the C-terminus of the Drozitumab heavy chain are binding similar or in a similar range to the human FAP expressed on the membranes of recombinant HEK cells as the IgG construct indicating that this fusion position does not have an N-terminal blocking effect on the tested FAP
binder. The used negative control molecules in this assay (secondary detection antibody and Drozitumab) do not bind in a detectable manner to FAP expressing HEK cells.
Table 4: Comparison of yield and quality of all different tested bispecific molecules.
-134-Molecule Yield Aggregates [(7(-1 Low molecular Monomer [rng/L1 before after SEC weight content [c7(-1 rc]
1 3F2 scFv_HC 2.53 59 0.00 4.13 96.34 (SEQ ID NO.:111) 2 4G8 scFv_HC 4.29 n.d. 0.00 1.50 98.50 (SEQ ID NO.:112) 3 4G8 scFv_LC 2.15 73 0.00 0.00 100.00 (SEQ ID NO.:113) 4 3F2 scFab_HC 3.99 72 0.00 0.00 100.00 (SEQ ID NO.:114) 3F2 scFab_LC 4.99 n.d. 0.00 0.00 100.00 (SEQ ID NO.:115) 6 4G8 scFab_HC 2.03 78 0.00 0.00 100.00 (SEQ ID NO.:116) 7 4G8 scFab_LC 2.47 66 0.00 0.00 100.00 (SEQ ID NO.:117) 8 4G8-X-Fab_A 36.10 90 0.00 0.00 100.00 (SEQ ID NO.:118, 119,120) 9 4G8-X-Fab_B 16.67 94 0.00 0.00 100.00 (SEQ ID NO.:121, 122, 119) In figure 8 the results of Surface Plasmon Resonance analysis (SPR, Biacore) are shown in which the simultaneous binding of the bispecific CrossFab molecules to DR5 and FAP was evaluated. For this assay the antigen (human DR5 as Fc fusion, SEQ ID NO.:316) was coupled 5 to the Biacore chip followed by injection of the CrossFab molecules as first analyte. After binding of the scFab molecules to DR5, recombinant soluble human or murine FAP
(SEQ ID
NO.:156 and 157) was used as the second analyte. For all tested bispecific molecules concentration dependent simultaneous binding to DR5 and human and murine FAP
was demonstrated, as indicated by the increase of the overall response rate after injection of the first analyte, obtained upon injection of the second analyte. Both tetravalent bispecific CrossFab
-135-molecules (2x2; Drozitumab-X-FAP_A and Drozitumab-X-FAP_B) showed similar binding to DR5 and human and murine FAP (figure 8; A-D).
Figure 9 shows the induction of apoptosis as determined by DNA fragmentation assay after 24 hrs of Drozitumab vs. two different bispecific Drozitumab¨X¨FAP molecules and one Drozitumab_4G8_scFab construct on the breast carcinoma cell line MDA-MB-231 in the absence or presence of FAP expressing fibroblast cells GM05389. Under the applied conditions, hyper-cross-linked Drozitumab and the bispecific DRS ¨ FAP molecules exhibit concentration dependent induction of apoptosis. While the bispecific constructs at low concentrations seemed to be dependent on the so-called bystander apoptosis (apoptosis activity only with DRS and FAP
expressing cell lines present) they also induced apoptosis of MDA-MB-231 cells alone at high concentrations. Hyper-cross-linked Drozitumab induced high levels of apoptosis also at low concentrations when only MDA-MB-231 cells were present. Both bispecific molecules exhibited maximal apoptosis induction already at a concentration of 0.7 nM.
To test the specificity of the bispecific constructs for their apoptosis induction activity being dependent on cross-linking via FAP a different assay set up was chosen (figure 10). In this setting recombinant human FAP or an unrelated control protein (white bars) were coated onto ELISA plates. These proteins were incubated with the bispecific molecules or the relevant controls before target cells (MDA-MB-231) were added and incubated for 24 hrs.
A
concentration dependent DNA fragmentation indicative for apoptosis induction could be demonstrated for the hyper-cross-linked Drozitumab (grey bars), a bispecific scFab molecule (hatched bars) and the bispecific CrossFab molecules (black bars) as shown in figure 10. While apoptosis induction with cross-linked Drozitumab always was in the same range with FAP or the control protein coated (over the entire tested concentration range) the apoptosis activity of CrossFab molecules was higher when FAP was coated onto the plates compared to the control protein. At concentrations of 0.7 nM and below significant apoptosis only could be detected in the presence of coated FAP, indicating specific cross-linking of DRS on the target MDA-MB-231 cells. The bispecific Drozitumab-FAP constructs containing the FAP moiety fused as a CrossFab to the C-terminus of the Drozitumab heavy chain exhibit superior apoptosis induction over Drozitumab alone at the tested concentrations for different tumor cell lines analyzed in co-culture apoptosis assays (DNA fragmentation assay) over 24 hrs as shown in figure 11. In this assay FAP expressing fibroblasts (GM05389) were co-cultured with a series of different tumor cell lines (MDA-MB-231, breast cancer, black bars; U-87MG, glioblastoma, grey bars; FaDu, head and neck, white bars and A549, lung cancer, hatched bars) in the presence of either Drozitumab, Drozitumab cross-linked with anti-Fc antibody, or Drozitumab-X-FAP
construct (all at concentrations of 7 nM and 0.7 nM). Apoptosis induction of cross-linked Drozitumab at a concentration of 7 nM was set to 100 % and the activities of the other tested molecules were calculated accordingly. While Drozitumab alone at a concentration of 7 nM
showed up to 50 %
-136-activity (depending on the tested tumor cell line), the CrossFab molecule exhibited apoptosis induction in the range of 75 ¨ 90 % of hyper cross-linked Drozitumab with all tested cell lines.
Drozitumab alone at 0.7 nM demonstrated low activity while the bispecific molecule displayed significant apoptosis induction activity (up to 100 % with FaDu cells, between 60 and 90 % with the other tested cell lines).
Example 4: Preparation of antigens and screening tools for the generation of novel DR5 binders Due to some suboptimal properties of Drozitumab in bispecific format (low productivity, active without cross-linking, N-terminal fusion inactivates the antibody) new DR5 antibodies were isolated which overcome these liabilities. For generation of suitable antigens to be used in isolation of novel DR5 binders and for screening and selection of those, a series of fusion proteins have been constructed. From each receptor gene the extracellular domain (ECD) encoding region was amplified by PCR and fused to a generic protein partner to generate the following formats (as depicted in figure 12):
1. Extracellular domain (ECD) fused in frame to Avi-tag and Hexahis-tag 2. ECD fused to Fc of human IgG1 consisting of the hinge region and CH2 and domains followed by an Avi tag. Between ECD and Fc an AcTev protease cleavage site was inserted (--(resulting in a dimeric antigen) 3. As in 2 but the ECD is fused to an Fc with knob-into-hole mutations. The antigen-Fc fusion is co-expressed with the Fc-hole counterpart to obtain monomeric antigens While the sequences of human DR5 and murine DRS were known and annotated in the SwissProt database, the sequence of the cynomolgus homolog has not been described there.
Based on homologies among human and rhesus DRS gene sequences primers have been designed that were used for isolation of the cynomolgus antigen from RNA
prepared from cynomolgus PBMCs. In brief, RNA was isolated from freshly isolated cynomolgus blood using the RNeasy Kit from Qiagen. After DNAseI digestion and elution of the RNA the OneStep RT-PCR kit (cDNA synthesis and amplification) from Qiagen (catalog number 210212) was used to amplify the cynomolgus DRS gene with GAB-4039 (GCTGGCTCCTGGACTTCCATTTCC, SEQ ID NO 163) and GAB4040 (GACCCAGGGAGGCGCGGGGAG; SEQ ID NO 164) as primers, designed according to the known rhesus DRS sequence. The PCR product was cloned into pCR2.1 Topo (Invitrogen) for sequencing. Analysis of the sequence revealed 89%
homology to the human DRS extracellular domain. Using the primers GAB-4145 (GTGCATTCCATCACCCGACAATCCCTAGATCCCCAGCG; SEQ ID NO 165) and GAB-
-137-4146 (GCGTCGACTGATTCTTTGTGGACACACTCAATGTCAC; SEQ ID NO 166) the extracellular domain of the cynomolgus DR5 gene was cloned into a generic expression vector in fusion with human Fc.
Expression of the desired genes occurs under control of a MPSV promoter. In addition a 3' polyadenylation site is included and an oriP sequence for stable maintenance of the plasmids in EBV nuclear antigen (EBNA) expressing HEI(293 cells.
For production of the antigens relevant expression vectors were transfected into HEI(293 EBNA cells (either Ca2PO4 mediated or PEI dependent transfection). After 5 ¨ 7 days of cultivation supernatants were harvested and purified via Protein A binding (Fc containing antigens) or by NI2+ affinity chromatography (Histag containing molecules) and subsequent size exclusion chromatography (SEC). If necessary the proteins were biotinylated via the C-terminal Avi tag. This could either be performed in vivo, by co-transfection / co-expression of a birA
encoding plasmid or after purification of the antigen in vitro using a biotinylation kit from Avidity Cat No. BIRA. The following antigens were produced by transient gene expression in HEI(293 EBNA:
Table 5: Antigen constructs and screening tools for isolation of antibodies against human DRS
I
# Antigen Amino Format I Source Ace. No acid 56 Open 1 human DR5 207 ECD ¨ Avi-his Biosystems 014763 BC001281.1 56 ¨
2 human DR5 ECD ¨ AcTev ¨ hu Fc ¨ Avi as above 014763 56¨ ECD ¨ AcTev ¨ hu Fc knob ¨
3 human DRSas above 014763 207 Avi Open 53 ¨ Q9QZM
4 murine DRS ECD ¨AcTev ¨ hu Fc ¨ Avi Biosystems BC065141.1 described herein cynom. 58 ¨ SEQ ID
5 ECD ¨ AcTev ¨ hu Fc ¨ Avi and WO
DRS 185 NO. 317 n.a.: not applicable Recombinant human DR4-Fc (Cat No. 347-DR/CF), DcRl-Fc (Cat No. 630-TR/CF), DcR2-Fc /Cat No. 633-TR/CF) and OPG-Fc (Cat No. 805-=S/CF) were purchased from R&D
Biosystems.
-138-Example 5: Isolation of novel anti DR5 binders from generic Fab libraries Antibodies with specificity for human DR5 were selected from a generic phage-displayed antibody library in the Fab format (DP47-3). This library was constructed on the basis of human germline genes using the V-domain pairing Vk3_20 (kappa light chain) and VH3_23 (heavy chain) comprising randomized sequence space in CDR3 of the light chain (L3) and CDR3 of the heavy chain (H3). Library generation was performed by assembly of 3 PCR-amplified fragments by splicing by overlapping extension (SOE) PCR. Fragment 1 comprises the the 5' end of the antibody gene including randomized L3, fragment 2 is a central constant fragment spanning from L3 to H3 whereas fragment 3 comprises randomized H3 and the 3' portion of the antibody gene.
The following primer combinations were used to generate these library fragments for DP47-3 library: fragment 1 (LMB3 ¨ LibLlb_new), fragment 2 (MS63 ¨ MS64) and fragment 3 (Lib2H
- fdseqlong), respectively. PCR parameters for production of library fragments were 5 min initial denaturation at 94 C, 25 cycles of 1 min 94 C, 1 min 58 C, 1 min 72 C and terminal elongation for 10 min at 72 C. For assembly PCR, using equimolar ratios of the 3 fragments as template, parameters were 3 min initial denaturation at 94 C and 5 cycles of 30 s 94 C, 1 min 58 C, 2 min 72 C. At this stage, outer primers were added and additional 20 cycles were performed prior to a terminal elongation for 10 min at 72 C. After assembly of sufficient amounts of full length randomized Fab constructs, they were digested NcoI I
Nod alongside with similarly treated acceptor phagemid vector. 22.8 ug of Fab library were ligated with 16.2 ug of phagemid vector. Purified ligations were used for 68 transformations to obtain a final library size of 4.2 x 101 . Phagemid particles displaying the Fab library were rescued and purified by PEG/NaC1 purification to be used for selections.
Selections were carried out against HEK293-expressed monomeric or dimeric human DR5 fused to the Fc-portion of a human IgG1 antibody. For the generation of the monomeric antigen, the Fc knob-into-holes format was applied for heterodimerization of two different CH2-CH3 chains (only one of which carrying human DR5 ectodomain as N-terminal fusion).
The antigens were enzymatically biotinylated via an avi-tag. Panning rounds were performed in solution according to the following pattern: 1. Preclearing of ¨ 1012 phagemid particles using hu IgG1 coated at 10 ug/m1 onto NUNC maxisorp plates to avoid Fc-binders, 2. binding of non-Fc binding phagemid particles from the supernatant of the pre-clearing reaction to 100 nM
biotinylated human DR5 for 0.5 h in a total volume of 1 ml, 3. capture of biotinylated hu DRS
and attached specifically binding phage by incubation on neutravidin-coated microtiter plates for 10 min, 4. washing of beads using 5x 1 ml PBS/Tween20 and 5x 1 ml PBS, 5.
elution of phage particles by addition of 1 ml 100 mM TEA (triethylamine) for 10 min and neutralization by addition of 500 n1 1M Tris/HC1 pH 7.4, 6. post-clearing step of eluted phage particles on human DcR2 to avoid cross-reactive binders and 7. re-infection of log-phase E. coli TG1 cells with the phage particles in the supernatant, infection with helperphage VCSM13 and subsequent
-139-PEG/NaC1 precipitation of phagemid particles to be used in subsequent selection rounds.
Selections were carried out over 3 rounds using constant antigen concentrations at 100nM. In round 2, capture of antigen: phage complexes was performed by addition of 5.4 x 107 streptavidin-coated magnetic beads for 10 min instead of capture on neutravidin-coated microtiter plates. Specific binders were identified by ELISA as follows: 100 1 of 50 nM
biotinylated human DR5 or DcR2 per well were coated on neutravidin plates.
Moreover, 10 ng/m1 human IgG1 were coated on NUNC maxisorp plates. Fab-containing bacterial supernatants were added and binding Fabs were detected via their Flag-tags by using an anti-Flag/HRP secondary antibody. Clones exhibiting signals on human DRS but none on human DcR2 and human IgG1 were short-listed for further analyses.
Affinity (KD) of selected Fab clones was measured by surface plasmon resonance using a ProteOn XPR36 instrument (Biorad) at 25 C with biotinylated mono- or bivalent DR5 antigens immobilized on NLC chips by neutravidin capture. Immobilization of antigens (ligand):
Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH
7.4, 0.005% Tween 20) to 10 ng/ml, then injected at 30 .t1/minute at varying contact times, to achieve immobilization levels of 200, 400 or 800 response units (RU) in vertical orientation.
Injection of analytes: For one-shot kinetics measurements, injection direction was changed to horizontal orientation, two-fold dilution series of purified Fab (varying concentration ranges between 100 and 3.125 nM) were injected simultaneously at 50, 60 or 100 1/min along separate channels 1-5, with association times of 120, 180 or 200s, and dissociation times of 200 or 240s.
Buffer (PBST) was injected along the sixth channel to provide an "in-line"
blank for referencing.
Association rate constants (kor,) and dissociation rate constants (kc,ff) were calculated using a simple one-to-one Langmuir binding model in ProteOn Manager v3.1 software by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) was calculated as the ratio koff/kõõ. Regeneration was performed in horizontal orientation using 50mM NaOH at a flow rate of 100 t1/min for a contact time of 18s.
Example 6: Selected DR5 binders are capable of inducing apoptosis upon cross-linking To identify DRS binders which are able to induce apoptosis of selected target cells only upon cross-linking the antibodies isolated from a Fab library were converted into the corresponding hu IgG1 format. In brief, the variable heavy and variable light chains of 46 unique DRS binders from phage display were amplified by standard PCR reactions using the Fab clones as the template. The PCR products were purified and inserted (either by restriction endonuclease and ligase based cloning, or by `recombineering' using the InFusion kit from Invitrogen) into suitable expression vectors in which they are fused to the appropriate human constant heavy or human constant light chain. The expression cassettes in these vectors consist of a chimeric
-140-MPSV promoter and a synthetic polyadenylation site. In addition, the plasmids contain the oriP
region from the Epstein Barr virus for the stable maintenance of the plasmids in HEK293 cells harboring the EBV nuclear antigen (EBNA). Antibodies were transiently produced in 50 ml scale in HEK293 EBNA cells as described. For a fast and high throughput purification, supernatants were neutralized and incubated with ProteinA Sepharose Fast Flow beads (GE
Healthcare Cat No. 17-5138-01) for 16h. The supernatant/bead mixture was then passed over an empty, equilibrated PD-10 column (GE Healthcare Cat No. 17-0435-01) by gravity flow. The retained beads were washed twice and the antibody eluted with a low pH step.
Finally, the eluted protein was neutralized and its concentration calculated using the asorbance at 280 nm and the molar extinction coefficient. The aggregate content of the antibody sample was analysed by analytical size exclusion chromatography using a Zorbax GF-250 column (Agilent Cat No PSMO 845006).
The results of this purification procedure are summarized in table 6 Table 6: Summary of purification results, SEQ ID NOs see Table 8 anti DR5 Titer Yield Monomer content mAb img/m11 i [mg/mil : [%]
3E6 47.3 32.9 100 4C3 43.6 21.2 100 5E11 38.2 29.9 100 5F6 40.4 38.0 100 6A8 41.9 28.1 100 6F8 64.4 45.1 99 7B12 75.1 44.6 100 7H9 54.7 38.5 99 18F11 77.6 54.4 100 18F12 66.6 33.4 99 18H6 54.6 48.5 100 19F10 61.1 36.7 100 20F2 65.9 41.0 99 21C3 51.4 40.6 97 212 53.6 27.1 99 23C7 49.8 27.2 100 24B7 38.5 28.9 100 1B12 179.3 78.1 100 2C5 116.2 48.7 98 2E5 144.3 67.7 100 2F11 146.2 67.3 100 4A6 148.0 83.2 100 4F5 110.5 48.7 100 4G9 128.1 80.9 100 6F6 161.3 86.3 100 6H4 128.1 65.8 100
-141-8D2 136.6 65.8 100 17A7 136.1 62.7 100 17H5 155.7 77.1 100 18A4 138.9 67.1 100 18A6 103.9 54.7 100 18Al2 118.1 63.3 100 18E6 90.8 44.3 100 18F6 99.3 36.0 100 19C12 192.6 70.6 92 19D6 144.9 81.7 97 19G7 117.0 63.1 100 20E3 116.4 51.7 100 20F1 75.5 40.7 100 21H3 127.1 45.1 98 22E6 130.9 61.2 98 22E9 137.5 60.2 100 23G10 127.9 43.5 100 6F9 96.8 48.2 100 6G7 114.7 59.3 100 17A8 102.1 60.0 100 This small scale transfection and production followed by purification via ProteinA beads yielded in reasonable amounts of pure antibodies with very low aggregate content. In figure 13 (a-e) the results of a standard DNA fragmentation ELISA assay for detection of apoptosis are summarized. Each antibody was tested in two different concentrations (1.0 ug/ml: black bars and 0.1 ug/ml: hatched bars with 1.0 and 0.1 ug/m1 of anti-Fc antibody for cross-linking, respectively). The obtained data were normalized to the maximal activity of cross-linked Drozitumab (at 1.0 OM) which was set to 100%. The majority of analyzed antibodies were able to induce concentration dependent induction of apoptosis in MDA-MB-231 cells after cross-linking. Seven of 46 binders (15.2 %) clearly showed higher activity as Drozitumab at both concentrations. Fifteen antibodies (32.6 %) were in the same activity range as Drozitumab and twenty of the tested molecules (43.5 %) did induce apoptosis upon cross-linking but to a much lower degree compared to Drozitumab. The remaining four binders ("20E3", "19E6", 1B12" and "19C12", 8.7 %) were inactive and did not induce apoptosis, even after cross-linking via a secondary anti Fc antibody.
Example 7: Characterization of novel DR5 binders Based on the results of apoptosis activity screen twelve of 46 evaluated DR5 antibodies were selected for additional, more detailed analysis and characterization.
These antibodies were re-produced in transiently transfected HEK293 EBNA cells and purified using standard ProteinA
-142-affinity columns followed by size exclusion chromatography as described. Yield and monomer /
aggregate content were determined (table 7). The purified antibodies were characterized with respect to target specificity, species cross-reactivity and affinity (table 8). In addition thermal stability was analyzed by DLS and the apoptosis induction activity was compared in the presence or absence of a secondary cross-linking anti-human Fc antibody.
Table 7: Series of selected DR5 antibodies, SEQ ID NOs see Table 8 SIX) 11) SEQ 11) Yield Monomer . . . Uross-An1ih dy N( ). NO. Spectfic11-1 i VI. .
I mg/LI I ( ( 1 reactvit VI I
specific for DR5 Human and 18F11 cynomolgous 94 95 33.00 100.0 monkey no binding to DR5, DcR1, no binding to DcR2 or OPG murine DRS
18H6 8.40 71.2 as above as above 18E6 31.50 100.0 as above as above 6H4 30.30 100.0 as above as above 5F6 42.18 100.0 as above as above 20F2 106 107 30.00 100.0 as above as above 4G9 42.90 100.0 as above as above 22E9 100 101 39.96 100.0 as above as above 21H3 102 103 32.35 100.0 as above as above
-143-4F5 36.63 100.0 as above as above 5E11 7 8 31.68 100.0 as above as above 24B7 19.73 100.0 as above as above Table 8: Characterization of novel DR5 binders ntibody SEQ ID NO. Affinity Avidity Aggregation temperature ,A
VH / VL ritM] [nM] [ C1 I
18F11 94/95 504 1.1 64 18H6 555 2.7 n.d.
18E6 471 2.2 66 6H4 773 8.9 65 5F6 552 1.5 66 20F2 106/107 431 4.8 66 4G9 478 6.4 65 22E9 100/101 217 1.5 64 21H3 102/103 259 1.6 64 4F5 575 4.4 n.d.
5E11 7/8 162 1.1 65 24B7 300 2.3 65 All analyzed antibodies specifically recognize human DRS and do not bind to the closest human homologs from the TNFR super family such as DR4, decoy receptors (DcR1 and DcR2) and osteoprotegerin (OPG). All DRS antibodies are cross-reactive with human and cynomolgus DRS but do not recognize the murine counterpart (which is not unexpected due to a sequence homology of only about 30%). While the affinities to human DRS are in a quite wide range (from 162 to 773 nM) all measured avidities are in the low (one digit) nanomolar range. All tested DRS binders reveal a high thermal stability with aggregation temperatures well above 60 C as measured by Dynamic Light Scattering (DLS) experiments.
To determine functional activity of a series of selected DRS binders, apoptosis induction was analyzed using a Cell Death Detection ELISA assay (Roche; #11 774 425 001) which specifically detects DNA fragmentation. Antibodies were used at a concentration of 1.0 and 0.1 ug/m1 in the presence of the same concentration of secondary anti human Fc antibody for cross-linking. For comparison antibodies were used at 1.0 ug/m1 in the absence of the secondary
-144-antibody to evaluate if the activity of the selected DR5 binders depends on cross-linking or if they are already active on their own. The human breast cancer cell line MDA-MB-231 was used as the target cell line. In figure 14 the results of the apoptosis induction of different DRS binders are summarized. After cross-linking all of the newly isolated DRS antibodies induce apoptosis in the same range as the control antibody Drozitumab. However, unlike Drozitumab which already exhibits significant apoptosis induction activity without cross-linking, activity of the novel DRS
binders strictly depends on cross-linking via a secondary antibody. The apoptotic activity does not seem to be correlated to the affinity of the DRS binder to its target since this was similar for antibodies with both the highest and the lowest affinity to human DRS.
Two additional assays were used to evaluate the activity of selected DRS
binders:
Inhibition of proliferation upon treatment with cross-linked antibodies was measured with a CellTiter-Glo assay (Promega #TB288). In addition, induction of Caspase 8 was determined by a Caspase8-Glo assay (Promega #G8202). Figure 15 shows the maximal inhibition of proliferation at a concentration of 7 nM of DRS binders cross-linked via secondary anti Fc antibody of three different tumor cell lines. The effect on the human colorectal adenocarcinoma cell line DLD-1 (black bars) was compared to the large cell lung cancer line HCI-H460 (hatched bars) and the human breast cancer cell line MDA-MB-231 (white bars). For all three cell lines significant inhibition of proliferation could be detected. While the inhibition of proliferation of DLD-1 at 7 nM seems to correlate with the affinity of the DRS antibody this is not the case for NCI-H460 and only to a certain degree for MDA-MB-231.
Table 9: Calculated IC50 values for inhibition of proliferation using the CellTiter-Glo assay Antiho(1. SEQ ID NO. I )1 D- 1 NCI-11460 M1).\-MR-231 NTH / V L
5E11 7/8 0.50 3.91 >7.00 22E9 100/101 0.56 4.00 >7.00 21H3 102/103 1.57 6.20 >7.00 24B7 1.86 5.00 > 7.00 20F2 106/107 2.01 6.53 >7.00 4G9 4.30 > 7.00 > 7.00 18E6 4.62 6.89 > 7.00 5F6 6.39 > 7.00 > 7.00 18F11 94/95 >7.00 >7.00 >7.00 4F5 >7.00 >7.00 >7.00 6H4 > 7.00 > 7.00 > 7.00 Figure 16 summarizes the results of Caspase 8 activation upon treatment with anti Fc cross-linked DRS antibodies. The maximal Caspase 8 activation in three different tumor cell lines (DLD-1, black bars; NCI-H460, hatched bars; and MDA-MB-231, white bars) is shown at a concentration of 7 nM. For all antibodies high level of Caspase 8 activation could be detected in all three cell lines. Induction of Caspase 8 was in the same range for the different cell lines but
-145-the correlation of Caspase 8 induction level with the affinity of the antibodies was not as pronounced as seen in the CellTiter-Glo Assay for inhibition of proliferation.
Table 10: Calculated EC50 values for induction of Caspase 8 using the Caspase 8-Glo assay Antibody SEQ ID NO. . DLI)-1 NCI-H460 MDA-MB-231 VH / VL
5E11 7/8 0.13 0.03 0.14 22E9 100/101 0.15 0.05 0.24 21H3 102/103 0.20 0.09 0.31 24B7 0.21 0.09 0.26 20F2 106/107 0.31 0.13 0.33 4G9 0.48 0.38 1.04 18E6 0.33 0.13 0.35 5F6 0.76 0.28 1.00 18F11 94/95 0.36 0.37 0.43 4F5 0.49 0.16 0.89 6H4 1.84 0.70 0.99 Example 8: Epitope analysis To characterize the kind and relative localization of the epitopes recognized by the newly isolated DRS binders in more detail, Western / Dot Blot analysis and Biacore measurements have been performed with purified IgGs. Comparison of Western Blot vs. Dot Blot results would show if the antibodies recognize a linear or a conformational epitope while Biacore competition experiments would hint to different, identical or partially overlapping epitopes.
To differentiate between linear or conformational epitopes human DRS was separated by SDS-PAGE, blotted onto a Nitrocellulose membrane, incubated with the different DRS binders and detected with a secondary anti-huFc-HRP antibody (Sigma A0170). In parallel human DRS
was spotted onto a membrane, DRS antibody was added and detected with the same secondary antibody. Binding of the antigen only in the Dot Blot experiment would hint to a conformational epitope since in this setting the antigen is analyzed in its natural three dimensional conformation whereas in the Western Blot experiment the antigen has been denatured and only linear epitopes should be accessible.
Table 11: Summary of results from Western / Dot Blot analysis Anti DR5 mAh SEQ ID NO. Simal in Vs/cstem Blot Si imal in Dot Blot VH / VL
4G9 +++
6H4 ++
-146-5F6 +++ +++
18F11 94/95 ++ +++
18H6 ++ +++
20F2 106/107 + +++
18E6 + +++
22E9 100/101 - ++
21H3 102/103 - +++
4F5 - ++
5E11 7/8 - +++
24B7 +++ +++
Based on these results it could be concluded that the majority of the analyzed antibodies recognize a conformational epitope on hu DR5 while only two antibodies (5F6, 24B7) clearly bind to the denatured antigen in Western Blot analysis indicating binding to a linear epitope.
Strong binding in Dot Blot and weak binding in Western Blot also hint to a conformational epitope but this probably contains linear stretches that are recognized (18F11, 18H6).
Binding competition assays by Surface Plasmon Resonance (SPR, Biacore) were performed in two different settings. In the classical sandwich assay a first DRS binder is immobilized on a chip followed by addition and binding of hu DRS. Then the second DRS
antibody is injected and analyzed for additional binding to DRS. In the tandem assay hu DRS is immobilized on a chip followed by addition of a first DRS binder. Then the second DRS binder is injected and additional binding is analyzed. Figure 17 summarizes the results of these binding competition assays assays (comparison of four new DRS antibodies with Drozitumab). From these competition assays it could be concluded that the clones 5E11, 22E9 and 174 (VH SEQ ID
NO.:88, VL SEQ ID NO. :89, see e.g. Example 26) probably share a common epitope since once one of these has been bound to DRS none of the two others could bind to the antigen. These three antibodies clearly recognize a different epitope as Drozitumab. The binding site of clone 422 (VH SEQ ID NO:82, VL SEQ ID NO. :85 see e.g. Example 26) might overlap with the epitope of 5E11, 22E9 and Drozitumab but definitely not with the one from clone 174. A clear answer to the question of the recognized epitope might come from co-crystallization experiments (DRS + mAb) followed by structure analysis.
To evaluate if the DRS antibodies recognize an epitope that is similar or overlaps with the DRS ligand binding site a TRAIL competition assay in Biacore was set up. For that purpose recombinant hu TRAIL (Preprotech No 310-04) was immobilized on a CMS chip. A
preformed complex consisting of human DRS-Fc and the DRS binder was used as analyte and binding to the immobilized TRAIL was determined. In figure 18 the results of the TRAIL
competition experiment are shown. With the exception of clone 422 (dotted line in figure 18b) all newly isolated DRS binders seem to bind to an epitope that at least overlaps with the TRAIL binding
-147-site on DR5 since none of the tested complexes is able to bind to immobilized hu TRAIL (all solid lines). In contrast to that, DR5 alone (dashed line) or a complex of DR5 with a control antibody (short dashes) showed clear binding to the immobilized ligand.
Example 9: Non-ligand competing DR5 antibodies demonstrate increased in vitro apoptosis activity in the presence of TRAIL compared to ligand competing binders During isolation and screening of new DR5 binders, antibodies were isolated which recognize different epitopes on human DRS. One possible criterion to classify the antibodies is to group them in ligand blocking and ligand non-blocking molecules. Among the selected binders one representative for each of these groups was chosen: clone 5E11 was identified to block TRAIL binding to DRS, while clone 422 is a ligand non-blocking antibody (figure 18).
To evaluate the impact of binding to an epitope that at least overlaps with TRAIL binding, these two candidates were analyzed for inhibition of proliferation of DLD-1 CRC target cells after cross-linking with secondary anti Fc antibodies in the presence or absence of human TRAIL. 4000 DLD-1 target cells per well were incubated over night at 37 C
before cross-linked antibodies were added: the antibodies 5E11, 422 and a control antibody that does not block TRAIL were used in six different concentrations (starting from 6.67 nM in 2.5 fold dilution steps down to 0.068 nM). Cross-linking was achieved by addition of equimolar concentration of anti Fc secondary antibody. TRAIL was added in concentrations of 10, 4, 1.6, 0.64, 0.256 and 0.102 nM to the respective antibody concentration. After 48 hrs incubation Cell TiterGlo reagent was added. The results of this growth inhibition assay are shown in figure 19. It was demonstrated that clone 422, the non-ligand blocking antibody exhibited significantly increased anti proliverative activity upon addition of human TRAIL compared to clone 5E11 which binds to a TRAIL binding site overlapping epitope. Addition of cross-linked 5E11 antibody had no additive effect on growth inhibition by TRAIL alone. In contrast, using cross-linked clone 422 in combination with human TRAIL showed a significant additive effect on activity.
Similar results were observed with a relevant control antibody known to not block TRAIL
binding on DRS. If this effect observed in an in vitro assay can be translated into in vivo settings remains to be evaluated.
Example 10: Conversion in bispecific 2+2 format In order to evaluate whether the novel DRS binders can be used for the generation of bispecific antibodies for the targeted induction of apoptosis of tumor cells by hyper-cross-linking of DRS, a set of DRS antibodies were converted into tetravalent bispecific molecules. These bispecific antibodies contain two binding moieties, each for DRS and FAP
(fibroblast activation protein). Different, selected DRS antibodies were combined with the FAP
antibody 28H1, a high affinity, human/ murine /cynomolgous monkey (hu/mu/cy) cross-reactive FAP
binder isolated
-148-and affinity matured by phage display. In the used 2+2 format the 28H1 CrossFab domain (VHCL) was fused to the C-terminus of the anti DR5 heavy chain using a (G4S)4 connector.
Schematic structures of the 2+2 format are shown in figure 28). Below are exemplary chains of the bispecific antibodies in the 2+2 format.
Table 12a: Bispecific, tetravalent DRS ¨ FAP CrossMab molecules (all with 28H1 CrossFab domain (VHCL) fused to the C-terminus of the anti DRS heavy chain using a (G4S)4 connector, FAP binder: VH SEQ ID NO.:15, VL SEQ ID NO.:16 ) DRS SEQ ID NO Name Description Binder VH /VL
(DRS) DRS (22E9)-28H1 28H1 CrossFab domain (VHCL) fused to the C-VHCL 2+2 terminus of the anti DRS (22E9) heavy chain using a (G45)4 connector:
VH (DR5)_Fc part ¨ VH (FAP) -CL chain (SEQ ID
NO.:125) VL (DRS)-kappa light chain (SEQ ID NO.:126) VLCH1 (FAP) chain (SEQ ID NO.:124).

DRS (21H3)-28H1 28H1 CrossFab domain (VHCL) fused to the C-VHCL 2+2 terminus of the anti DRS (21H3) heavy chain using a (G45)4 connector:
VH (DR5)_Fc part ¨ VH (FAP) -CL chain (SEQ ID
NO.:125) VL (DRS)-kappa light chain (SEQ ID NO.:128) VLCH1 (FAP) chain (SEQ ID NO.:124).

DRS (20F2)-28H1 28H1 CrossFab domain (VHCL) fused to the C-VHCL 2+2 terminus of the anti DRS (20F2) heavy chain using a (G45)4 connector:
VH (DR5)_Fc part ¨ VH (FAP) -CL chain (SEQ ID
NO.:129) VL (DRS)-kappa light chain (SEQ ID NO.:130) VLCH1 (FAP) chain (SEQ ID NO.:124).
-149-5E11 7/8 DR5(5E11)-28H1 28H1 CrossFab domain (VHCL) fused to the C-VHCL 2+2 terminus of the anti DRS (5E11) heavy chain using a (G4S)4 connector:
VH (DR5)_Fc part ¨ VH (FAP) -CL chain (SEQ ID
NO.:131) VL (DRS)-kappa light chain (SEQ ID NO.:132) VLCH1 (FAP) chain (SEQ ID NO.:124).
5E11 7/8 DR5(5E11)-28H1 As above, and removal of C-term. Lysine and VHCL 2+2 P329G/LALA mutation in Fc VH (DR5)_Fc part ¨ VH (FAP) -CL chain (SEQ ID
NO.:134) VL (DRS)-kappa light chain (SEQ ID NO.:132) VLCH1 (FAP) chain (SEQ ID NO.:124).
18F11 94/95 DR5(18F11)-28H1 28H1 CrossFab domain (VHCL) fused to the C-VHCL terminus of the anti DRS (18F11) heavy chain 2+2 using a (G45)4 connector:
VH (DR5)_Fc part ¨ VH (FAP) -CL chain (SEQ ID
NO.:140) VL (DRS)-kappa light chain (SEQ ID NO.:141) VLCH1 (FAP) chain (SEQ ID NO.:124).
Table 12b: Production data of bispecific, tetravalent DRS ¨ FAP CrossMab molecules D R5 binder Production yield Final monomer Apoptosis induction [mg/L1 %I
5E11 20.3 96.7 +++
22E9 18.1 99.0 +++
21H3 7.6 96.3 +++
20F2 19.3 100.0 +++
18E6 25.1 100.0 ++
5F6 18.9 99.4 18F11 22.3 96.7 6H4 18.5 100.0 ++
18H6 17.5 99.6
-150-All DR5 ¨ FAP bispecific molecules were produced in transiently transfected EBNA cells and were purified via Protein A and size exclusion chromatography.
The obtained product yields were in a reasonable range (around 20 mg/L). The monomer content after the final purification step was above 96 % for all molecules.
Target binding analysis by surface plasmon resonance (Biacore) revealed that all selected bispecific antibodies in the 2+2 format were able to simultaneously bind to recombinant DRS
and FAP (human and murine) as depicted in figure 42.
To evaluate if the DRS-FAP bispecific molecules are able to induce apoptosis of the MDA-MB-231 target cell line, 96 well plates were coated with recombinant human FAP
for cross-linking of DRS on the target cells via the subsequently added bispecific antibodies. After addition of the target cells (MDA-MB 231) and incubation for 24 hrs apoptosis induction was determined by the standard DNA fragmentation ELISA assay. Figure 20 shows the comparison of apoptosis induction of seven bispecific antibodies containing newly isolated DRS binders and the C-terminally fused 28H1 FAP CrossFab compared to Drozitumab at two different concentrations (7.0 nM and 0.7 nM). All molecules were tested in the presence and absence of FAP. As expected, at high concentration the Drozitumab based bispecific molecule induced apoptosis already in the absence of cross-linking FAP to a significant degree.
In contrast, the DRS-FAP bispecific molecules using the new DRS binders only exhibited apoptosis induction activity in the presence of FAP coated on the plates indicating that this activity is dependent on the cross-linking via recombinant FAP.
To determine if the bispecific molecules also could be efficiently cross-linked via FAP
expressed on a different cell line than DRS and thereby induce apoptosis, a so-called bystander co-culture assay was set up. Figure 21 shows the results of apoptosis induction in a tumor cell line (MDA-MB-231) and FAP expressing fibroblast (GM05389) co-culture experiment with three different concentrations of bispecific molecules (7.0, 0.7 and 0.07 nM).
The three bispecific constructs containing the DRS binding moieties from 5E11, 22E9 and 20F2, respectively fused to the 28H1 FAP CrossFab show a comparable degree of induction of apoptosis as it also was observed with the corresponding, anti Fc cross-linked IgG molecules. In contrast to that the molecule containing 6H4 as the DRS binding part exhibited reduced activity compared to cross-linked IgG. One of the tested molecules (18H6 ¨ 28H1) was completely inactive at all concentrations (in contrast to the cross-linked IgG), indicating that not all DRS
binders are suitable for generation of bispecific molecules to hyper-cross-link DRS on tumor cells. Therefore a careful evaluation of epitope and bispecific activity is necessary to choose the right DRS antibody for the approach of targeted induction of apoptosis. In this regard DRS
binders which display a rather low affinity to DRS and a high avidity in the 2+2 bispecific format are particularly advantageous. The low affinity for DRS prevents binding of the bispecific
-151-antibodies to normal cells and hence increases the selectivity for inducing apoptosis in tumor cells in a FAP dependent manner. One binder having these characteristics is for example the DR5 binder 5E11.
As described, one of the disadvantages of Drozitumab, which limits its use in bispecific formats, is the fact that this antibody does not allow for N-terminal fusion of a second binding moiety. This configuration leads to the blocking of Drozitumab' s N-terminus which inhibits proper binding to DRS and thereby induction of apoptosis is prevented. To test if the newly isolated DRS antibodies can be used in this kind of format, Fc-fusion molecules were generated, in which the Fabs of selected DRS binders were fused C-terminally to a human Fc region via a (G4S)4 connector. These molecules were transiently produced in HEK293 EBNA
cells, purified via ProteinA beads and tested in an apoptosis induction assay. In figure 22 a) the results of the DNA fragmentation assay in MDA-MB-231 cells with these Fc-DR5 fusion molecules after cross-linking with secondary anti Fc IgG are summarized. All tested molecules are able to induce apoptosis of the target cell line, indicating that the chosen DRS binders are not N-terminally blocked which opens a wider range of formats that can be used with these binders.
Example 11: Characterization of thermal stability of 2+2 bispecific format containing phage display derived DR5 binder Thermal stability of phage display derived DRS binder converted into the 2+2 bispecific format was measured using an Optiml 000 system (Avacta Group plc) as the change in scattered light intensity. In a micro cuvette array, 9 p L of the samples in 20 mM Histidine, 140 mM NaC1, pH
6.0 at a concentration of 1 mg/mL were heated from 25 C to 90 C at a rate of 0.1 C/min.
Scattered light intensity (266 nm laser) was recorded every 0.4 C and processed with the software IgorPro, Version 6.23 (Avacta Group plc). The aggregation onset temperature is defined as the temperature at which the scattering intensity starts to increase.
Table 13: Aggregation onset temperatures of bispecific antibodies (2+2 format) containing phage display derived DRS binders as a measure for thermal stability of the bispecific constructs TheraPS Name Alias Bispec SEQ ID NOs Optim1000 Bispec Antibody Tagg ( C) Antibody DR5TAA-0030 5E11-28H1 131,132, 124 64 DR5TAA-0037 22E9_28H1 124,125,126 63 DR5TAA-0038 21H3_28H1 127 ,128,124 63 DR5TAA-0039 20F2 28H1 129,130,124 64
-152-All tested DR5 antibodies reveal a high thermal stability with aggregation temperatures well above 60 C also when cloned into 2+2 bispecific constucts.
Example 12: DR5 ¨ FAP bispecific antibodies are able to induce apoptosis on different target cells Nine DRS ¨ FAP bispecific antibodies in the 2+2 format comprising newly isolated DRS
binders (four isolated by phage display, five derived from rabbit immunization as depicted in examples 22 to 25) fused to the FAP 28H1 CrossFab moiety were compared in a side-by-side experiment for induction of apoptosis on two different cell lines (MDA-MB-231 and G401) in a co-culture assay with GM05389 human FAP fibroblasts. The bispecific antibodies were tested over a concentration range from 0.0007 ¨ 7 nM. The results of the DNA
fragmentation assay are summarized in figure 23 (MDA-MB-231) and figure 24 (G401). All bispecific antibodies tested demonstrated good apoptosis induction activity on both cell lines. According to the results obtained with the MDA-MB-231 cells the antibodies could be divided into two groups. The one obtained from rabbit immunization showed the maximum of apoptosis induction at a concentration of 0.7 nM and with further increasing antibody concentration the activity decreased slightly. In contrast, the phage display derived binders in the bispecific 2+2 format did not show the decline in activity at high concentrations but stayed constant or even more increased up to the highest concentration. In the experiment with G401 cell in co-culture with GM05389 fibroblasts for both sets of bispecific antibodies the maximum of apoptosis induction was reached already at a concentration of 0.07 nM and then stayed constant. In this setting all molecules performed similarly in terms of apoptosis induction levels.
Example 13: Comparison of different bispecific formats It has been demonstrated that DR5-FAP bispecific molecules in the 2+2 CrossFab format produce well in a very good quality and are able to mediate concentration-dependent specific induction of apoptosis in a two cell line co-culture setting. The degree of apoptosis induction of these bispecific molecules is in the same range as observed with the corresponding DRS binders that were hyper-cross-linked via a secondary anti Fc antibody. To evaluate additional, different DR5-FAP bispecific molecules for their apoptosis induction capacity, the following constructs have been generated in which DRS and FAP binding entities are combined in different formats and with different valences as shown in figure 25.
Table 14: Description of alternative DR5-FAP bispecific formats Description Valency DR5 FAP
-153-Fusion of FAP binder as CrossFab (VHCL) to the C-terminus of 2+1 5E11 DRS heavy chain (knob). B24_E11A_001 DR5(5E11)-28H1Fc knob VHCL 2+1 (SEQ ID NO. 142) DR5(5E11) Fc hole (SEQ ID NO. 143) FAP (28H1) VLCH1 (SEQ ID NO. 124) DRS(5E11) LC (SEQ ID NO. 132) Comment: Knob-into-hole; (G45)4 connector, Figure 25a 2 FAP CrossFab (VHCL) on Fc-hole. DRS Fabs (head-to-tail) 2+1 5E11 fused to Fc-knob. B24_E1 1E_001 DR5(5E11)_Fc knob Fab-Fab Head-to-tail 2+1 (SEQ ID NO.
145) FAP (28H1)_Fc holeVHCL (SEQ ID NO. 146) FAP (28H1) VLCH1 (SEQ ID NO. 124) DRS(5E11) LC (SEQ ID NO. 132) Comment: Knob-into-hole;(G45)2 connector, Figure 25b 3 FAP CrossFab (VHCL) fused to the C-terminus of DRS Fc-hole. 3+1 5E11 DRS Fab fused to C-terminus of DRS Fc-knob. B24_E11A_002 DR5(5E11)-28H1 Fc knobVHCL 3+1 (SEQ ID NO. 143) DRS(5E11)-DRS(5E11) Fc hole 3+1(SEQ ID NO. 144) FAP (28H1) VLCH1 (SEQ ID NO. 124) DRS(5E11) LC (SEQ ID NO. 132) Comment: Knob-into-hole; (G45)4 connector, Figure 25c 4 Fusion of FAP binder as CrossFab (VHCL) to the C-terminus of 2+1 18F11 DRS heavy chain (knob). B16_E11A_001 DR5(18F11)-28H1Fc knobVHCL 2+1 (SEQ ID NO. 147) DRS(18F11) Fc hole (SEQ ID NO. 148) FAP (28H1) VLCH1 (SEQ ID NO. 124) DRS(18F11) LC (SEQ ID NO. 141) Comment: Knob-into-hole; (G45)4 connector, Figure 25a FAP CrossFab (VHCL) fused to the C-terminus of DRS Fc-hole. 3+1 18F11 28H1 DRS fused to C-terminus of DRS Fc-knob. B16_EllA_002 DR5(18F11)-28H1VHCLFc knob 3+1 (SEQ ID NO. 149) DR5(18F11)-DR5(18F11) Fc hole (SEQ ID NO. 150) FAP (28H1) VLCH1 (SEQ ID NO. 124) DRS(18F11) LC (SEQ ID NO. 141) Comment: Knob-into-hole; (G45)4 connector, Figure 25c
-154-All molecules were transiently produced in HEK293 EBNA cells and were purified according to standard Protein A and size exclusion chromatography protocols.
Side product profile and quality of the molecules were analyzed by SDS-PAGE, SEC and Caliper analysis.
Table 15: Summary of production results of additional bispecific DR5-FAP
molecules Construct I )escription Yield Aggregate Monomer Low molecular [mg/nil sI ( ( I (I weight species 1 B24_E1 1 E_00 5E11_28H1; 5.42 1.55 95.2 3.25 1 2+1 C-terminal on Fc-knob 2 B24_E1 1A_00 5E11_28H1; 5.99 0.74 98.08 0.74 1 head-to-tail on Fc-knob 3 B24_E1 1A_00 5E11_28H1; 5.39 1.80 98.20 0.00 2 3+1 C-terminal on Fc-knob 4 B16_E11A_00 5E11_28H1; 18.34 1.50 98.50 0.00 1 2+1 C-terminal on Fc-knob B16_E11A_00 5E11_28H1; 3.32 0.90 99.10 0.00 2 3+1 C-terminal on Fc-knob As summarized in table 15 these additional formats seem to be more difficult to be produced and purified compared to the 2+2 formats. Except one (B16_El 1A_001), all show lower expression yields as the analogous 2+2 molecules. However, the aggregate contents were rather low (< 2%) but in most cases also lower molecular weight species could be detected which might be due to purification or degradation of the molecules.
Apoptosis induction activity on MDA-MB-231 was tested in the fibroblast co-culture assay (DNA fragmentation) at different concentrations (7.0; 0.7 and 0.07 nM). Figure 26 shows the results of apoptosis activity of three different formats (2+2; 2+1 and 3+1) compared to bispecific Drozitumab in the 2+2 format. In this setting the 5E11-28H1 bispecific molecule in the 2+2 format overall showed similar activity compared to the Drozitumab control. At the lowest concentration, the 5E11-28H1 bispecific molecule in the 2+2 format had displayed even higher activity compared to the Drozitumab control. In contrast, the 2+1 and the 3+1 formats seemed to be less active compared to both 2+2 formats (Drozitumab and 5E11). If the same formats are analyzed with a different DRS binder (18F11) all three molecules show reduced apoptosis induction activity as compared to the Drozitumab bispecific antibody. However, with the 18F11
-155-bispecific molecule the maximal apoptosis induction occurred at lower concentrations (0.07 nM) whereas the Drozitumab based molecule exhibits higher activity with increasing concentration.
In a second set of molecules an additional 2+1 format was evaluated for the bispecific antibody in which two DRS binding moieties are fused head-to-tail to an Fc-knob part while the FAP targeting Fab is combined with an Fc-hole counterpart. All four formats were compared over a broad range of concentrations (ranging from 7.0 to 0.0007 nM) in a DNA
fragmentation assay for apoptosis induction of MDA-MB-231 in co-culture with fibroblasts (figure 27). Both 2+1 molecules had the highest apoptosis activity at a concentration of 0.7 nM while the 3+1 molecule showed maximal induction of apoptosis already at a 10 fold lower concentration. However, none of the new formats seemed to be superior over the conventional 2+2 format.
Example 14: Test of functional activity of a 1+1 bispecific format in a co-culture assay system Besides the different format and valency variants described in example 12, another construct has been generated in a 1+1 format (as depicted in fig. 25 D) with the DRS binder 0011 (= clone 174) in the uncrossed Fab and the FAP binder 4B9 in the crossed Fab (SEQ ID NOs.
280-282). Functional activity of the 1+1 format was compared to a 2+2 format (as depicted in fig.
28 A, containing 0011 as DRS binder and 28H1 as FAP binder; SEQ ID NOs 287-289) and to the chimeric IgG of 0011 (SEQ ID NOs 90-93) with or without secondary crosslinking.
Apoptosis induction was measured by Cell Death Detection ELISA (CDDE, Roche #11 774 425 001) upon treatment of cells with anti-DRS and anti-DRS-FAP bispecific antibodies in the presence or absence of a cross-linking secondary antibody in a co-culture system consisting of tumor cells (MDA-MB-231) and fibroblasts (GM05389).
Day 1: Preparation of cells. The adherent MDA-MB-231 cell line (human breast adenocarcinoma) was grown in DMEM medium (PAN) supplemented with 10% fetal calf serum (Gibco) and 2mM L-glutamine (PAA), and normally split twice per week 1:20. The GM05389 fibroblasts were grown in MEM + Earle' s medium (Gibco) supplemented with 15% fetal calf serum (Gibco), lx NEAS (PAN) and 2mM L-glutamine (PAA), and normally split twice per week 1:3.
For co-culture assays fibroblast were seeded on day 1 at a density of 1x104 cells/100p1/well in 96-well plates and incubated overnight at 37 C, 5% CO2. Tumor cells and antibodies were added on day 2.
-156-For monoculture assays cell lines were seeded on different plates at the same density, incubated overnight and treated with antibodies the next day.
Day 2: Induction of apoptosis. Medium was aspirated and antibodies (anti-DR5 IgGs or anti-DRS-FAP bispecific antibodies) were added to the cells at different final concentrations (see figures) alone or together with the cross-linking antibody (goat anti-human IgG, Sigma #I2136) in a 1:1 ratio in 100p1 medium.
For co-culture assays tumor cells were immediately added (1x104 cells/100pl/well) on the fibroblasts after the antibodies to reach a final volume of 100p1.
Cultures were incubated at 37 C for 24 hs.
Day 3: Cell Death Detection Elisa (CDDE). The immunoassay was performed according to the manufacturer's instructions (Roche) with slight changes. Briefly, cells were lysed with 100p1/we11 2x-lyse buffer for 15 minutes at RT. A master mix consisting of anti-histone and anti-DNA antibodies was prepared according to the manufacturer's instructions and mixed with 1:4 diluted lysates on 96-well streptavidin-coated flat-bottomed microtiter plates (Roche). After a 2-hour incubation at RT, wells were washed, ABTS substrate added and incubated at RT until color development sufficient for photometric analysis (10-30 min). Absorbance was read at 405nm with a Tecan Spectra Rainbow Reader.
Results are shown in Figures 46 and 47.
While the monospecific anti-DRS antibodies (both chimeric 0011 and Drozitumab) did not induce significant apoptosis by their own, hyperclustering of the DRS
molecules on the cell surface with a secondary antibody led to cell death at concentrations around 0.7 mM. However, best apoptosis induction was achieved when anti-DRS-FAP bispecific antibodies (in either format 1+1 or 2+2) were added to the co-culture leading to DRS hyperclustering on the tumor cells via the second moiety which binds to FAP present on the surface of fibroblasts (figure 46).
While the monospecific anti-DRS antibodies induced apoptosis of MDA-MB-231 cells upon cross-linking, bispecific molecules against DRS and FAP were only functional in the presence of DRS-expressing tumor cells and FAP-expressing tumor associated fibroblasts. In addition, none of the molecules showed any effect on the viability of fibroblasts. (Relative apoptosis as compared to internal assay positive control, see figure 47).
This activity data demonstrates that FAP specific apoptosis induction can also be achieved with a DRS-FAP bispecific antibody in a 1+1 format with one valency for each target in a comparable efficacy as with a 2+2 format.
-157-Example 15: The 2+2 bispecific format as a generic platform technology The bispecific DR5-FAP CrossFab molecules in the 2+2 format in which the 28H1 VHCL
is fused to the C-terminus of the 5E11 heavy chain has been proven to be a very good format.
For different DRS binders it has demonstrated to be producible at reasonable product titers, it is stable, exhibits only low amounts of missing or wrongly paired light chains and reproducibly shows good activity. Nevertheless, to extend the format platform, additional bispecific 5E11-28H1 molecules have been generated which differ in the kind and location of the crossing point.
The five additional formats evaluated and the parental molecule are depicted in figure 28. Four of the five additional molecules do not contain a crossed 28H1 Fab anymore but a standard Fab domain fused to the C-terminus of the DRS (5E11) heavy chain (either VHCH1 or VLCL which is fused by a (G4S)4 connector).
Table 16: Description of additional DRS-FAP CrossFab molecules Fi2. 28 scetch C 1) 1 1 Kind of Crossing N-terminal N-terminal N-terminal C-terminal C-terminal Fab VHVL CH1CL Fab VHVL
5E11 IgG VLCL ¨ Fc VLCH1 ¨ VHCL ¨ VHCH1 ¨ VHCH1 ¨ Fc Fc Fc Fc 28G1 Fab (fusion VHCH1 VHCH1 VHCH1 VLCL VLCH1 to heavy chain C-terminus) Table 17: Exemplary sequences of additional DRS-FAP CrossFab molecules VLCL Anti FAP 28H1 Fab (VHCH1) fused to the C-terminus of the DRS (5E11)_28H1 DRS (5E11) heavy chain by a (G4S)4 connector with entire 2+2 Fab crossed (VLCL-Fc) (molecule B in Fig 28) VLCL (DRS) ¨ Fc ¨ VHCH1 (FAP) (Seq ID NO. 274) VHCH1 (DRS) (SEQ ID NO. 275) VLCL (FAP 28H1) (SEQ ID NO. 137) VHVL Anti FAP 28H1 Fab (VHCH1) fused to the C-terminus of the DRS(5E11)-28H1 DRS (5E11) heavy chain by a (G45)4 connector with crossing 2+2 in Fab (VLCH1-Fc) (molecule C in Fig 28) VL (DRS)-CH1- Fc part-VH(FAP)-CH1 chain (SEQ ID NO.
135) VH(DR5) ¨CL ( SEQ ID NO.:136)
-158-VL (FAP) ¨kappa light chain (SEQ ID NO.:137).
Anti FAP 28H1 Fab (VHCH1) fused to the C-terminus of the CH1CL DR5 (5E11) heavy chain by a (G4S)4 connector with crossing in Fab (VHCL1-Fc) DRS(5E11)-28H1 VH (DRS) CL- Fc- -VH(FAP) -CH1 chain (SEQ ID
2+2 NO.:138) (molecule D in Fig 28) VL(DRS) ¨CH1 chains (SEQ ID NO.:139) VL (FAP) ¨kappa light chains (SEQ ID NO.:137) Anti FAP (28H1) Fab (VLCL) fused by a (G45)4 connector to DRS (5E11)-28H1 C-terminus of DRS (5E11) Fc (VLCL) VH (DRS)-CH1-Fc-VL (FAP)-CL (SEQ ID NO. 276) 2+2 VL (DRS)-CL (SEQ ID NO. 132) (molecule E in Fig 28) VH (FAP)-CH1 (SEQ ID NO. 277) Anti FAP (28H1) CrossFab (VLCH1) fused by a (G45)4 DRS (5E11)-28H1 connector to the C-terminus of DRS (5E11) Fc (VLCH1) VH (DRS)-CH1-Fc-VL (FAP)-CL (SEQ ID NO.278) 2+2 VL (DRS)-CL (SEQ ID NO. 132) (molecule F in Fig 28) VH (FAP)-CL (SEQ ID NO. 279) All molecules were transiently produced in 200 ml scale in HEK293 EBNA cells, purified by standard ProteinA and size exclusion chromatography and finally analyzed and characterized in comparison with the original format (C-terminal CH1CL crossing of 28H1 fused to 5E11 heavy chain, (molecule A, fig 28)).
As summarized in table 16 the five molecules can be divided into two groups:
one contains the two formats in which the entire Fab's are crossed (B + E). These two molecules gave very low product titers and even after purification they still contained high amounts of aggregate. Therefore these two molecules were not further evaluated. The other three (crossing of 5E11 VHVL or CH1CL and crossing of the C-terminal 28H1 as VHVL) exhibited much better product quality with respect to yield and aggregate content. The molecules C, and D also were tested for target binding on MDA-MB-231 cells by FACS in comparison to molecule A as shown in figure 29. Furthermore functionaly activity in terms of apoptosis induction, simultaneous and independent target binding, stability and aggregation tendency as well as side product patterns of molecules C, D, and F were compared to format A (Examples 22 to 25)
-159-Example 16: Test of functional activity of 2+2 bispecific format variants in a coculture assay system The four different 2+2 format variants (format A, C, D, F of fig 28) were compared for their functional activity to induce apoptosis as measured by Cell Death Detection ELISA (CDDE, Roche #11 774 425 001) upon treatment of cells with the constructs in a co-culture system consisting of tumor cells (MDA-MB-231) and fibroblasts (GM05389).
Day 1: Preparation of cells. The adherent MDA-MB-231 cell line (human breast adenocarcinoma) was grown in DMEM medium (PAN) supplemented with 10% fetal calf serum (Gibco) and 2mM L-glutamine (PAA), and normally split twice per week 1:20. The GM05389 fibroblasts were grown in MEM + Earle's medium (Gibco) supplemented with 15% fetal calf serum (Gibco), lx NEAS (PAN) and 2mM L-glutamine (PAA), and normally split twice per week 1:3.
For co-culture assays fibroblast and tumor cells were seeded on the same day each at a density of 1x104 cells/100pl/well in 96-well plates and incubated overnight at 37 C, 5%
CO2. For monoculture assays cell lines were seeded on different plates at the same density.
Day 2: Induction of apoptosis. Medium was aspirated and bispecific antibodies were added to the cells at different final concentrations (see figures) in 100p1 medium.
Cultures were incubated at 37 C for 24 hs.
Day 3: Cell Death Detection Elisa (CDDE). The immunoassay was performed according to the manufacturer's instructions (Roche) with slight changes. Briefly, cells were lysed with 100p1/we11 2x-lyse buffer for 15 minutes at RT. A master mix consisting of anti-histone and anti-DNA antibodies was prepared according to the manufacturer's instructions and mixed with 1:4 diluted lysates on 96-well streptavidin-coated flat-bottomed microtiter plates (Roche). After a 2-hour incubation at RT, wells were washed, ABTS substrate added and incubated at RT until color development sufficient for photometric analysis (10-30 min). Absorbance was read at 405nm with a Tecan Spectra Rainbow Reader.
Results are shown in Figure 30. Induction of apoptosis of 4 different CrossMab variants in co- (a) and mono-culture (b, c) settings as detected by DNA fragmentation. All 4 different variants induce apoptosis in tumor cells in the co-culture setting in a comparable dose-dependent manner.
In mono-culture settings no apoptosis is induced neither in MDA-MB231 tumor cell line nor in GM05389 fibroblast cell line, pointing out the specificity and FAP-dependency of apoptosis
-160-induction of all 4 variants. This functional activity data demonstrates that the bispecific 2+2 format can be used in different configurations with respect to type and site of crossing.
Example 17: Test of independent and simultaneous target binding of 2+2 bispecific format variants The four different 2+2 format variants (format A, C, D, F of fig 28) were compared additionally for their ability to bind both targets independently and simultaneously by SPR
assays.
Assessment of independent DR5- and FAP-binding to different Crossmab Variants Around 3000 resonance units (RU) of the capturing system (5 p g/ml anti human IgG (Fc); GE
Healthcare, BR-1008-39) were coupled on a CM5 chip (GE Healthcare BR-1005-30) at pH 5.0 by using an amine coupling kit supplied by the GE Healthcare. The sample and system buffer was PBS-T (10 mM phosphate buffered saline including 0.05% Tween20) pH 7.4.
The temperature of the flow cell was set to 25 C and of the sample block to 12 C. Before capturing, the flow cell was primed with running buffer twice.
The bispecific antibody was captured by injecting a 5p g/ml solution for 60 sec at a flow of 5 t1/min. Independent binding of each ligand to the bispecific antibody was analysed by determining the active binding capacity for each ligand, either added sequentially or simultaneously (flow of 10 t1/min):
1. Injection of human DR5 with a concentration of 5p g/ml for 180 sec (identifies the single binding of the antigen).
2. Injection of human FAP with a concentration of 5p g/ml for 180 sec (identifies single binding of the antigen).
3. Injection of human DRS with a concentration of 5p g/ml and of human FAP
with a concentration of 5p g/ml for 180 sec (identifies the binding of DRS and of FAP
at the same time).
The surface was regenerated by 60 sec washing with a 3m MgC12 solution at a flow rate of 30 t1/min. Bulk refractive index differences were corrected by subtracting the response obtained from an anti human IgG surface.
The bispecific antibody is able to bind both antigens mutual independently if the resulting final signal of the approach 3 equals the sum of the individual final signals of the approaches 1 and 2.
-161-Table 18: Quantitative Assessment of the independent DR5- and FAP-binding of 4 different Crossmab variants SEQ ID NO DR5 FAP expected measured ratio signal signal Mix:
expected DR5 + FAP DR5 + FAP
measured [RU [RU [RU max ] [RU max ]
max ] max ]
DR5TAA- 134,132,124 37,1 40,8 77,9 76,7 98 format A
DR5TAA- 135,136,137 33,0 41,9 74,9 73,1 98 format C
DR5TAA- 138,139,137 27,1 38,4 65,5 63,6 97 format D
DR5TAA- 278,132,279 26.7 7.2 34 33 97 format F
All 4 different Crossmab variants are able to bind DRS & FAP mutually independent.
Assessment of simultaneous DR5- and FAP-binding to the Crossmab First, around 600 resonance units (RU) of DR5 (20p g/m1) were coupled on a CM5 chip (GE Healthcare BR-1005-30) at pH 5.0 by using an amine coupling kit supplied by the GE
Healthcare. The sample and system buffer was PBS-T (10 mM phosphate buffered saline including 0.05% Tween 20) pH 7.4. Flow cell was set to 25 C and sample block to 12 C and primed with running buffer twice. Second, 10 p g/ml solution of the bispecific antibody was injected for 30 sec at a flow of 30 t1/min. Third, hFAP (10p g/m1) was injected for 30 sec at a flow of 30 t1/min. The binding response of hFAP depends from the amount of the bispecific antibody bound to hDR5 and shows simultaneous binding. The surface was regenerated by 70 sec washing with a Glycine pH2 solution (GE Healthcare BR-1003-55) at a flow rate of 30 t1/min. Simultaneous binding is shown by an additional specific binding signal of hFAP to the previous DRS bound Crossmab.
-162-Assessment of the simultaneous DR5- and FAP-binding of four Crossmab formats (DR5TAA-0057, DR5TAA-0078, DR5TAA-0077 and DR5TAA-0081) showed that all 4 different Crossmab variants are able to bind FAP and DRS simultaneously.
Example 18: Test of thermal stability and aggregation tendency of 2+2 bispecific format variants The four different 2+2 format variants (format A, C, D, F of fig 28) were furthermore analyzed for their thermal stability and for their tendency to form aggregates.
Thermal Stability Aggregation onset temperature: Samples were prepared at a concentration of 1 mg/mL in 20 mM
Histidine/Histidine chloride, 140 mM NaC1, pH 6.0, transferred into an optical 384-well plate by centrifugation through a 0.4 p m filter plate and covered with paraffin oil.
The hydrodynamic radius is measured repeatedly by dynamic light scattering while the samples are heated with a rate of 0.05 C/min from 25 C to 80 C. The aggregation onset temperature is defined as the temperature at which the hydrodynamic radius starts to increase.
Aggregation tendency Samples were dialyzed into formulation buffer (20 mM His/HisCl, 240 mM
Trehalose, pH 6.0) and adjusted to a concentration of 1 mg/mL. After sterile filtration over 0.22 p m centrifugal filter devices (Millipore), samples were stored for 2 weeks at 40 C, while a control sample was maintained at -80 C. Aggregate formation was monitored by SE-HPLC using a column (Tosoh) and reported as the difference between the 40 C and the -80 C
sample.
Table 19: Assessment of the thermal stability and aggregation tendency of 4 different Crossmab formats.

0081(5EQ ID
(SEQ ID NOs (SEQ ID NOs (SEQ ID NOs 134,132,124) 135, 136, 137) 138, 139, 137) 279) Format A Format C Format D
Format F
Stability by DLS
o 64 61 60 61 [Tagg, C]
Aggregate formation during storage @ 40 C in 0.4 0.3 2.2 0.5 formulation buffer [% increase]
-163-Thermal stability was slightly reduced in the formats C, D and F as compared to the parental format A but with 61 C and 60 C still in a good range.
Aggregation tendency was very low for formats A, C and F and slightly increased for format D.
Example 19: Evaluation of side-product profile of 2+2 bispecific format variants by mass spectrometry Finally, the four different 2+2 format variants (format A, C, D, F of fig 28) were furthermore analyzed for their side-product profile by mass spectrometry.
The deglycosylated total mass of the different constructs was determined and confirmed via electrospray ionization mass spectrometry (ESI-MS). Moreover potential side products such as LC overrepresentation were detected and relatively quantified. Briefly, 100 p g purified antibodies at a protein concentration of up to 3 mg/ml were deglycosylated with 14 U N-Glycosidase F (Roche) in 100 mM NaH2PO4/Na2HPO4, pH 7 at 37 C for 2 h and subsequently desalted via HPLC on a Sephadex G25 column (GE Healthcare). The deglycosylated total mass was determined via ESI-MS on a maXis UHR-TOF (Bruker) MS system equipped with a TriVersa NanoMate (Advion) source.
The different DRS-FAP bispecific antibody constructs were analyzed by mass spectrometry in their deglycosylated form to evaluate the product identity and integrity. The identity could be confirmed for all evaluated constructs. Moreover the different constructs showed several side products for example with overrepresentation of one LC type or with loss of one or two LCs. All constructs had qualitatively and quantitatively similar byproducts profiles.
All results from examples 13 and 30-33 celarly show that the bispecific 2+2 format can be used in different configurations with respect to type and site of crossing. This makes the 2+2 CrossFab format a broadly applicable platform technology.
Example 20: Drozitumab-FAP bispecific molecules exhibit superior in vivo efficacy over untargeted Drozitumab It has been demonstrated in in vitro activity assays that bispecific Drozitumab-FAP exhibit superior apoptosis induction activity compared to Drozitumab alone which is not cross-linked.
To evaluate if this also translates into in vivo efficacy in relevant mouse tumor models different xenograft models have been set up. One problem with the standard mouse tumor models is that the FAP expression usually is very low and does not reflect the human situation at all. To
-164-establish models that more closely resemble the FAP expression in human tumor stroma, engineered murine fibroblasts (3T3 cells) that recombinantly express mouse FAP
were co-injected with the respective tumor cell lines (colorectal carcinoma cell line DLD-1 in nude mice and breast cancer line MDA-MB-231 in SCID mice). For this purpose 3T3 fibroblasts were stably transfected with a plasmid that carries an expression cassette for the full length murine FAP gene under control of the MPSV promoter. For selection of stable clones the vector further carries an additional expression cassette for a puromycin acetyltransferase which confers resistance to puromycin. Several clones expressing different levels of murine FAP as judged by FACS binding experiments have been selected. One selected mu FAP-3T3 cell line was co-injected with tumor cell line DLD-1 leading to significantly enhanced tumor growth as compared to DLD-1 cells injected without mu FAP expressing 3T3 cells. Also, IHC
analysis using human /
mouse cross-reactive anti FAP antibodies has demonstrated high levels of FAP
expression in the tumor surrounding stroma for both cell lines, as expected. Therefore this co-grafted subcutaneous xenograft model was used to assess the in vivo efficacy of bispecific Drozitumab-FAP molecule compared to the original untargeted Drozitumab antibody. 2x106 tumor cells were co-implanted with 20 % of 3T3 fibroblast expressing murine FAP. Twelve (DLD-1) and ten days (MDA-MB-231) after tumor cell implantation therapy was started. The animals were injected (i.v.) either buffer, Drozitumab (10 mg / kg) or Drozitumab-FAP (10 mg / kg).
To compensate for the molecular weight difference of Drozitumab vs. bispecific Drozitumab (Drozitumab is only 60 % of the molecular mass of the bispecific molecule) the latter was administered twice weekly while Drozitumab was only given once a week. In figure 31 the increase of mean tumor volumes in mm3 over time are summarized. Figure 31 A shows in vivo efficacy of Drozitumab-FAP compared to Drozitumab and the buffer control in the DLD-1 / mu FAP-3T3 co-injection model. While Drozitumab only demonstrated a moderate anti-tumor efficacy, resulting in a tumor growth inhibition (TGI) of 36 % compared to the control, the bispecific molecule exhibited a tumor growth inhibition of 99 % at the end of the study (day 33).
In the MDA-MB-231 / mu FAP-3T3 co-injection model this difference is even more pronounced since in this model Drozitumab alone did not show any efficacy better than the buffer control whereas the Drozitumab-FAP bispecific antibody exhibited a tumor growth inhibition of 77 %
at the end of the study at day 32 (figure 31 B). This result might indicate that the MDA-MB-231 cell line is more resistant to apoptosis induction as the DLD-1 cell line.
Example 21: Evaluation of anti-tumor activity of DR5-FAP bispecific molecules using newly isolated DR5 binders in combination with 28111 FAP CrossFab.
In vivo efficacy of three different DR5 binders (5E11, 174 and 422) in bispecific format fused to the anti FAP antibody 28H1 as CrossFab was compared side-by-side in the DLD-1 and MDA-MB-231 xenograft models each co-injected with 3T3 fibroblast expressing murine FAP. A
-165-fourth molecule consisting of 5E11-28H1 CrossFab with mutations in the Fc region that completely abolish binding to Fcy receptors (while affinity to FcRn is unchanged) was included.
For this purpose large scale transient transfections and productions were conducted to generate sufficient material in HEK cells.
Table 20: Production of bispecific antibodies for in vivo experiments. All materials exhibited acceptable endotoxin content of < 0.23 EU / mg.
Sample Fc region Titer fmg/L I Yield [mg] Aggregate [%]
DR5(5E11)-1 FAP(28H1)VHCL wt 60 75 < 2 2+2 DR5(5E11)-2 FAP(28H1)VHCL PG_LALA* 80 100 < 2 2+2 DR5(174)-3 FAP(28H1)VHCL wt n.d. 65 < 1 2+2 DR5(422)-4 FAP(28H1)VHCL wt n.d. 96 < 1 2+2 L234A; L235A; P329G
All molecules were produced at good yields with excellent quality with respect to aggregate and endotoxin content. Binding to the relevant antigens was analyzed by different methods (SPR and FRET) as summarized in table 21.
Table 21 a: Affinities / Avidities of DR5-FAP bispecific molecules to human and cynomolgus antigens Sample Fc region Affinity [iiNIP Avidity [111µ11': Avidity [nM]+
hu cv DR5 hu cv DR5 hu cy DR5 1 wt 146.0 9.6 0.16 0.29 1.2 0.4 DR5(5E11) FAP(28H1) VHCL 2+2
-166-2 PG_LAL
147.0 11.9 0.14 0.26 1.9 0.6 DR5(5E11) A
FAP(28H1) VHCL 2+2 3 wt 9.8 20.2 0.10 3.72 1.1 0.3 DRS(174)-FAP(28H1) VHCL 2+2 4 DRS (422)- wt 5.1 2.0 0.08 n.d. 0.8 0.7 FAP(28H1) VHCL 2+2 * Biacore measurements TagLite Before in vivo experiments were initiated the materials were first tested for in vitro apoptosis induction activity. Figure 32 summarizes the results of a Cell Death Detection ELISA
in which four different DR5-FAP bispecific molecules were compared at concentrations of 7.0, 0.7 and 0.07 nM. The assay was set up as a co-culture assay in which DLD-1 cells were used as targeted cells and 3T3 or recombinant 3T3 cells expressing murine FAP served as the effector cells for cross-linking. In the DLD-1 ¨ 3T3 co-culture experiment hardly any induction of apoptosis was detectable in DLD-1 cells whereas in the setting with the FAP
expressing 3T3 cells a 10 fold increase in apoptosis induction was observed indicating that this activity is due to the cross-linking via FAP on the surface of the recombinant 3T3 cells. A
similar experiment was performed in which the same bispecific molecules, target and effector cells were used to determine cell viability upon treatment with the bispecific agonistic DR5-FAP
antibodies. The results of this experiment are summarized in figure 33. Here, a significant reduction of cell viability of DLD-1 cells only was observed in the presence of FAP expressing 3T3 cells while with unmodified 3T3 cells (which do not express murine FAP) no reduction of viability was seen.
All four bispecific DR5-FAP molecules were used for evaluation of in vivo efficacy in two different tumor models, both co-injected with 20 % of murine FAP expressing 3T3 fibroblasts.
The results of these in vivo efficacy experiments are shown in figure 34.
After engraftment of the tumor and fibroblast cells treatment started with 10 mg / kg administered twice weekly intravenously (i.v.). Definitely, all four molecules were able to control tumor growth in both models as demonstrated by significant tumor growth inhibition (TGI) compared to the vehicle control. The absolute percentage of tumor growth inhibition at the end of the study was in a similar range for all four molecules. The following results were obtained for the DLD-1 and MDA-MB-231 model, respectively: 5E11_28H1 (wt Fc): 75 % / 95 %; 5E11_28H1 (PGLALA):
83 % / 99 %; clone 174: 66 % / 87 % and clone 422: 73 % / 89 %. All molecules were slightly more potent in the MDA-MB-231 model than in the DLD-1 experiment.
-167-Table 21 b): Summary of characteristics of preferred DR5-FAP bispecific antibody Clone Name 5E11 (VH: SEQ ID NO.: 7 28H1 (VH: SEQ ID NO.: 15 VL: SEQ ID NO.:8) VL: SEQ ID NO.: 16) Affinity human 165 (IgG) 2.6 (IgG) ILnMI
Affinity Cyno 1.02 (IgG) 3.7 (IgG) ILnMI
Avidity Human 0.06 (IgG) 0.25 (IgG) Avidity Cyno 0.06 (IgG) 0.06 (IgG) Binding Mode agonistic (only upon No interference with cros slinking) signaling/protease function, TRAIL competitive, conformational epitope conformational epitope Specificity No binding to huDR4, No binding to hu DPP-IV
DcR1/2, OPG (CD26, closest FAP
homologue) Species Cross- Human, cyno Human, cyno, murine Reactivity Examples 22 to 25: Generation and characterization of new DR5 binding moieties by immunization Besides selection of new DR5 antibodies with improved properties from a phage display library (as described above) new DR5 antibodies were generated also by immunization of rabbits (examples 22-25) followed by intensive characterization (examples 26-28).
Example 22: Immunization of rabbits One set of rabbits was immunized with 400 p g of recombinant human DR5 (monomeric Fc fusion), emulsified with complete Freund' s adjuvant, at day 0 by intradermal application, and with 200 p g each of DR5-huFc, emulsified with complete Freund' s adjuvant, at days 7, 14, 35, 63 and 91, by alternating intramuscular and subcutaneous applications. Blood (10% of estimated total blood volume) was taken at days 21, 41, 69 and 97. Serum was prepared, which was used for titer determination by ELISA (see below), and peripheral mononuclear cells were isolated, which were used as a source of antigen-specific B cells in the B cell cloning process (Example 17).
Another set of rabbits was immunized genetically, using a plasmid expression vector coding for human DRS lacking the intracellular death domain, by intradermal application of 400 p g vector DNA, followed by Electroporation (5 square pulses of 750 V/cm, duration 10 ms, interval 1 s). Rabbits received 6 consecutive immunizations at days 0, 14, 28, 49, 77 and 105.
-168-Blood (10% of estimated total blood volume) was taken at days 35, 56, 84 and 112. Serum was prepared, which was used for titer determination by ELISA (see below), and peripheral mononuclear cells were isolated, which were used as a source of antigen-specific B cells in the B
cell cloning process (Example 23).
Determination of serum titers Human DR5 (monomeric Fc fusion), was immobilized on a 96-well NUNC Maxisorp plate at 0.3125 pg/ml, 100 p 1/well, in PBS, followed by blocking of the plate with 2% Crotein C in PBS, 200 p 1/well; application of serial dilutions of antisera, in duplicates, in 0.5% Crotein C in PBS, 100 p 1/well; detection with HRP-conjugated donkey anti-rabbit IgG
antibody (Jackson Immunoresearch) diluted 1:16 000 in 0.5% Crotein C in PBS, 100 p 1/well. For all steps, plates were incubated for 1 h at 37 C. Between all steps, plates were washed 3 times with 0.05%
Tween 20 in PBS. Signal was developed by addition of BM Blue POD Substrate soluble (Roche), 100 p 1/well; and stopped by addition of 1 M HC1, 100 p 1/well. Absorbance was read out at 450 nm, against 690 nm as reference. Titer was defined as dilution of antisera resulting in half-maximal signal.
Example 23: B-Cell Cloning from Rabbits Isolation of rabbit peripheral blood mononuclear cells (PBMC) Three rabbits (described in the Example "Immunization of rabbits") were used as a source of blood. EDTA containing whole blood was diluted twofold with lx PBS (PAA, Pasching, Austria) before density centrifugation using lympholyte mammal (Cedarlane Laboratories, Burlington, Ontario, Canada) according to the specifications of the manufacturer. The PBMCs were washed twice with lx PBS.
EL-4 B5 medium RPMI 1640 (Pan Biotech, Aidenbach, Germany) supplemented with 10% FCS
(Hyclone, Logan, UT, USA), 2mM Glutamin, 1% penicillin/streptomycin solution (PAA, Pasching, Austria), 2mM sodium pyruvate, 10mM HEPES (PAN Biotech, Aidenbach, Germany) and 0,05 mM b-mercaptoethanole (Gibco, Paisley, Scotland) Depletion of macrophages/monocytes Sterile 6-well plates (cell culture grade) were used to deplete macrophages and monocytes through unspecific adhesion. Each well was filled at maximum with 4 ml medium and up to 6x106 PBMCs from the immunized rabbit and allowed to bind for 1 h at 37 C in the incubator.
The cells in the supernatant (peripheral blood lymphocytes (PBLs)) were used for the antigen panning step.
-169-Coating of plates For panning on protein sterile streptavidin coated 6-well plates (Microcoat, Bernried, Germany) were coated with 2 tg/m1 biotinylated recombinant human DR5 (monomeric Fc fusion) in PBS for 3 h at room temperature. For panning on human surface DR5-positive cells G401 cells were seeded in sterile cell culture 6-well plates and cultivated to generate a confluent cell monolayer. Prior to the panning these 6-well plates were washed with sterile PBS three times.
Enrichment of B cells on the human DR5 protein 6-well tissue culture plates coated with human DRS protein or covered with human DR5-positive G401 cells were seeded with up to 6x106 PBLs per 4 ml medium and allowed to bind for 1 h at 37 C in the incubator. After the enrichment step on the DRS antigen non-adherent cells were removed by carefully washing the wells 1-2 times with 1 x PBS. The remaining sticky cells were detached by trypsin for 10 min at 37 C in the incubator. Trypsination was stopped with EL-4 B5 medium. The cells were kept on ice until the immune fluorescence staining.
Immune fluorescence staining and Flow Cytometry The anti-IgG FITC (AbD Serotec, Dtisseldorf, Germany) was used for single cell sorting.
For surface staining, cells from the depletion and enrichment step were incubated with the anti-IgG FITC antibody in PBS and incubated for 45 min in the dark at 4 C. After staining the PBMCs were washed two fold with ice cold PBS. Finally the PBMCs were resuspended in ice cold PBS and immediately subjected to the FACS analyses. Propidium iodide in a concentration of 5 tg/m1 (BD Pharmingen, San Diego, CA, USA) was added prior to the FACS
analyses to discriminate between dead and live cells. A Becton Dickinson FACSAria equipped with a computer and the FACSDiva software (BD Biosciences, USA) were used for single cell sort.
B-cell cultivation The cultivation of the rabbit B cells was prepared by a method similar to that described by Zubler et al. (1985). Briefly, single sorted rabbit B cells were incubated in 96-well plates with 200 n1/well EL-4 B5 medium containing Pansorbin Cells (1:100000) (Calbiochem (Merck), Darmstadt, Deutschland), 5% rabbit thymocyte supernatant (charge TSN-M13 (10242), MicroCoat, Bernried, Germany) and gamma-irradiated murine EL-4-B5 thymoma cells (2,5 x 104/well) for 7 days at 37 C in an atmosphere of 5% CO2 in the incubator. The supernatants of the B-cell cultivation were removed for screening and the remaining cells were harvested immediately and were frozen at ¨ 80 C in 100 n1 RLT buffer (Qiagen, Hilden, Germany).
Example 24: B-cell PCR and Recombinant Expression
-170-Isolation of ribonucleic acid (RNA) The cells from which the RNA had to be isolated were at first pelleted by centrifugation.
The cell pellet was lysed by the addition of 100 1 RLT-buffer with 10 1/m1 beta-mercaptoethanol. The cells were resuspended by multiple mixing with a pipette and transferred to a multi well plate. The plate was shortly centrifugated at 200 x g and frozen at -20 C. The isolation of the RNA was performed with the NucleoSpinC) 96 RNA kit (Macherey & Nagel) according to the manufacturer's instructions.
Reverse transcription polymerase chain reaction The reverse transcription was carried out with SuperScript III First-Strand Synthesis SuperMix (Invitrogen) according to the manufacturer's instructions.
Polymerase chain reaction The polymerase chain reaction was carried out with AccuPrime Pfx SuperMix (Invitrogen) according to the manufacturer's instructions. Light chain and heavy chain variable regions were amplified in separate reactions. PCR-primers were used with 25 bp overlaps to target antibody expression vectors. PCR-products were purified by NucleoSpinC) 96 Extract II
kit (Macherey &
Nagel).
Sequencing and SLIC Cloning The PCR products were sequenced to determine the DNA-sequences of the variable regions of heavy and light chains. The PCR-products were cloned into expression vectors by the so called SLIC-cloning method, which is described by Haun, R.S., et al., in BioTechniques 13 (1992) pp. 515-518 and Li, M.Z., et al., in Nature Methods 4 (2007) pp. 251-256. The plasmids for the antibody expression were linearized by restriction anzyme digestion.
The linearized plasmids were purified by preparative agarose electrophoresis and extracted from the gel (Qiaquick Gel Extraction Kit / Qiagen). The purified plasmids were added to a PCR-protocol using overlapping primers (bay 25 bp) for the PCR-product to be cloned. Both the vector and insert were treated with T4 DNA polymerase (Roche Applied Sciences) in the absence of dNTPs to generate overhangs, then vector and insert were incubated with RecA (New England Biolabs) protein and ATP to promote recombination. Products were transformed into E.coli. Plasmid DNAs for light chain and heavy chains were isolated and each couple was combined for transient transfections.
Transient Transfection for Antibody Expression in HEK293 cells HEK293 cells (Invitrogen) were grown in F17-media (Gibco) to 1 x 10e6 cells/ml. 2 x 10e6 HEK293 cells were transfected with 1 p g HC + LC plasmids suspended in 293-free (Novagen) and OptiMEMC) (Gibco). After 7 days incubation supernatants were harvested, purified via Protein A and analyzed.
-171-Example 25: Screening of DR5 antibodies derived from immunization B-cell culture supernatants were screened by multiple parallel ELISA-based binding assays in 384 well microtiter plates. Antibodies binding to the DR5-expressing cells G401 and to biotinylated recombinant human DRS (monomeric Fc fusion) and cynomolgus DRS
(dimeric Fc fusion), but not to human DR4 (TNFRSF10A; R&D Systems Cat.No. 347-DR-100) and human IgG1 , were selected as primary hits. In a secondary screening, micropurified antibodies recombinantly expressed in HEK cells were tested again for binding to human and cynomolgus DRS and additionally for absence of binding to human DR4, human DcR1 (inhouse), human Dc R2 (in house) or human Osteoprotegerin (OPG; R&D Systems Cat.No. 805-05-100).
Furthermore antibodies were tested in a functional apoptosis assay (Cell Death Detection Elisa) on G401 cells in the presence or absence of a cross-linking anti-human Fc antibody. Only those antibodies able to induce apoptosis upon Fc-mediated cross-linking (but not in its absence) were selected for further characterization and development.
Sequences of antibodies selected after secondary screening and cloned in expression plasmids by the SLIC cloning procedure were verified by subcloning and re-sequencing of the variable light and variable heavy chains.
These subcloned and sequence-verified expression plasmids were then used for larger scale transient transfections of HEK293F cells followed by Protein A purification allowing further more intensified characterization steps.
Example 26: Characterization of binding properties of DR5 antibodies derived from immunization Selected DRS antibodies from rabbit immunization were characterized for their binding properties, species cross-reactivity and specificity by binding ELISA and SPR
analysis.
Binding of monoclonal antibodies to TRAIL binding receptors (Immunoassay) Antigen binding immunoassays were performed at room temperature on 384 well streptavidin coated microtiter plates (MicroCoat Biotechnologie GmbH) with PBS
buffer supplemented with 0.05% Tween -20 and 0.5 % BSA (Roche Diagnostics GmbH). 125 ng/ml biotinylated human DRS (monomeric Fc fusion) protein (inhouse) or 63 ng/ml biotinylated hFc cynomolgus DRS protein (inhouse) or 63 ng/ml biotinylated DcR2 (TNFRSF10D) protein (inhouse) were added to the wells containing a mixture of 1:3000 diluted anti-rabbit Fc¨HRP
conjugate (GE Healthcare) and 1:50 diluted B-cell supernatants. After 90 min incubation the plate was washed 6 times with PBST (phosphate buffered saline with 0.2%Tween -20) and developed with BM blue HRP substrate solution (BM blue : 3,3 '-5,5'-Tetramethylbenzidine,
-172-Roche Diagnostics GmbH) for 30 minutes at RT. Absorbance was measured at 370 nm. The blank value was defined without addition of supernatant.
For negative selection against the hFc tag of the immunogen an immunoassay with a mixture of biotinylated anti-human IgG (Fab specific) from Jackson ImmunoResearch (Cat. No.
109-066-006), human IgG1 (inhouse) and anti-rabbit Fc¨HRP conjugate was used and processed as described. For testing binding to related Trail binding receptors like human DR4 (TNFRSF10A; R&D Systems Cat.No. 347-DR-100), human Osteoprotegerin (OPG; R&D
Systems Cat.No. 805-05-100) and human DcR 1 (TNFRSF10C; R&D Systems Cat.No.

100) an immunoassay was established by capturing the respective protein ¨ hFc chimera with anti-human Fc antibody (Jackson ImmunoResearch, Cat. No. 109-006-098) on a MaxiSorp 384 well microtiter plate (Sigma-Aldrich, Nunc).
Table 22: Binding of DR5 antibodies (rabbit IgG) derived by immunization to human DRS
(monomeric Fc Fusion) and cynomolgus DRS as detected by biochemical ELISA
(EC50 [lig/mil). No significant binding to mouse DRS, human DR4, DcR1, DcR2, and Osteoprotegerin was detected (data not shown) DR5 Antibody DR5 EC50 EC50 SEQ ID
Clone Name Antibody human DR5 cynomolgus NO
Clone [ng/m1] DRS VH / VL
Alias [ng/m1]
DR5TAA-0005 039 2,5 3,7 41/46 DR5TAA-0006 058 2,0 1,6 51/55 DR5TAA-0010 481 2,8 8,7 60/64 DR5TAA-0013 298 2,1 2,1 68/71 DR5TAA-0019 461 3,5 2,2 74/78 DR5TAA-0016 422 4,0 2,7 82/85 DR5TAA-0011 174 2,0 4,3 88/89 Selected DRS antibodies from rabbit immunization show good and comparable binding properties to human and cynomolgus DRS in ELISA while they do not recognize murine DRS.
All selected antibodies are highly specific for DRS as they did not give significant signals in binding ELISA to human DR4, DcR1, DcR2, and Osteoprotegerin.
DRS kinetic affinity Around 3000-5000 resonance units (RU) of the capturing system (10 tg/m1 goat anti rabbit; ordering code JIR111-005-046; Jackson Immuno Research) were coupled on a CMS chip (GE Healthcare BR-1005-30) at pH 5.0 by using an amine coupling kit supplied by the GE
Healthcare. The sample and system buffer was PBS-T (10 mM phosphate buffered saline including 0.05% Tween20) pH 7.4. The flow cell was set to 25 C - and the sample block set to 12 C - and primed with running buffer twice.
-173-The DR5 antibodies were captured by injecting a 1 p g/ml solution for 30 sec at a flow of t1/min. Association was measured by injection of recombinant human DR5 (monomeric His-Avi fusion protein, in house) in various concentrations in solution for 120 sec at a flow of 30 t1/min starting with 100 nM down to 0.41 nM in 1:3 dilutions. The dissociation phase was 5 monitored for up to 300 sec and triggered by switching from the sample solution to running buffer. The surface was regenerated by 60 sec washing with a 100mM H3PO4 (phosphoric acid) solution at a flow rate of 30 t1/min. Bulk refractive index differences were corrected by subtracting the response obtained from a blank surface. Buffer injections are also subtracted (=
double referencing). For calculation of KD and other kinetic parameters the Langmuir 1:1 model 10 was used.
Table 23: Kinetic affinities of DR5 antibodies (rabbit IgG) derived by immunization DR5 Antibody DR5 Antibody ka kd KD SEQ ID NO
Clone Name Clone Alias [nM] [nM] [nM] VH / VL
DR5TAA-0005 039 1,60E+06 2,14E-02 1,33E-08 41/46 DR5TAA-0006 058 3,12E+05 1,85E-03 5,92E-09 51/55 DR5TAA-0010 481 1,11E+06 1,77E-02 1,59E-08 60/64 DR5TAA-0011 174 4,55E+05 1,93E-03 4,24E-09 88/89 DR5TAA-0013 298 4,07E+05 3,71E-03 9,12E-09 68/71 DR5TAA-0016 422 3,46E+05 5,02E-04 1,45E-09 82/85 DR5TAA-0019 461 9,00E+05 6,07E-04 6,75E-10 74/78 Example 27: Functional characterization of DR5 antibodies derived from immunization DNA fragmentation and cell viability DR5 antibodies were functionally characterized by evaluating apoptosis and cell viability as measured by Cell Death Detection ELISA (CDDE, Roche #11 774 425 001) and Cell Titer Glo (CTG, Promega #G7573), respectively, upon treatment with anti-DR5 antibodies in the presence or absence of a cross-linking antibody.
Preparation of cells: the adherent MDA-MB-231 cell line (human breast adenocarcinoma) was grown in DMEM medium (PAN) supplemented with 10% fetal calf serum (Gibco) and 2mM L-glutamine (PAA), and normally split twice per week 1:10. For the assay, cells were washed with PBS, detached from the flask with Accutase (PAA), seeded in 96-well flat-bottomed microtiter plates (Costar) at a density of 1x104 cells/well (CDDE) or 0.25x104 cells/well (CTG) in 50p1 and incubated overnight at 37 C, 5% CO2.
Induction of apoptosis by rabbit anti-DR5 antibodies: samples (rabbit anti-DR5 IgGs, DR5-TAA-#) were added to the cells at different concentrations alone or together with the cross-linking antibody (goat anti-rabbit IgG, Jackson Immunoresearch) in a 1:1 ratio in 50p1 in PBS, to
-174-induce crosslinking of the DR5 receptors leading to apoptosis. Cells were incubated at 37 C for 24 hs (CDDE) or 48hs (CTG).
A) Cell Death Detection Elisa (CDDE): the immunoassay was performed according to the manufacturer's instructions (Roche). Briefly, supernatants were carefully aspirated and cells lysed with 200 p1/well lyse buffer for 30 minutes at RT. A master mix consisting of anti-histone and anti-DNA antibodies was prepared according to the manufacturer' s instructions and mixed with the 1:4-diluted lysates on 96-well streptavidin-coated flat-bottomed microtiter plates (Roche). After a 2-hour incubation at RT, wells were washed, ABTS substrate added and incubated at RT until color development sufficient for photometric analysis (10-30 min).
Absorbance was read at 405nm with a Tecan Spectra Rainbow Reader.
Figure 35 shows the results of this experiment: The generated rabbit anti-DRS
antibodies were able to induce apoptosis of MDA-MB231 cells with different potencies but always in a dose-dependent fashion and only after Fc-mediated cross-linking of the DRS
molecules. In the absence of an anti-rabbit Fc-specific secondary antibody, no significant cell death was detected.
B) Cell Titer Glo (CTG): the immunoassay was performed according to the manufacturer' s instructions (Promega). Briefly, cells were first lysed in the buffer containing the luminescence substrate (100 1). After a 10-minute incubation period on a shaker luminescence was measured with the TECAN Infinite Plus reader.
Figure 36 shows the results of this experiment: Cell viability is diminished by anti-DRS
antibodies upon receptor hyperclustering in a dose-dependent manner. The generated rabbit anti-DRS antibodies were able to decrease the viability of MDA-MB231 cells with varying potencies but always in a dose-dependent fashion and only after Fc-mediated cross-linking of the DRS
molecules. In the absence of an anti-rabbit Fc-specific secondary antibody, the cell viability was not affected at any antibody concentration.
Cell viability & Caspase 8 activation Caspase8-Glo Caspase 8 activation assays (Promega cat#G8202) and CellTiter-Glo cell viability assays (Promega cat#TB288) were carried out according to the manufacture's instructions.
For Caspase 8 activation assays, 10,000 cancer cells were seeded in 75 pi per well in opaque white 96 well plates (BD Falcon cat#BD353296) and incubated at 37 C
with 5% CO2 overnight. Then anti-DRS antibodies were added together with anti-rabbitFc antibodies (equal molar concentration of DRS and rabbitFc antibodies in 25 1) in 6 serial dilutions. Antibodies were incubated on the cells at 37 C with 5% CO2 for 3 hours. Then 100 pi of caspase 8 substrate in lysis buffer was added to each well and mixed well. After incubation of another 30 minutes at 37 C luminescence signal was read in a Spectra Max M5 plate.
For Cell Viability assays, 4,000 cancer cells were seeded per well in black/clear bottom 96 well plates (BD Falcon cat#BD353220) and incubated at 37 C with 5% CO2 overnight. Then
-175-anti-DR5 antibodies were added together with anti-rabbitFc antibodies (equal molar concentration of DR5 and rabbitFc antibodies in 25 1) in 6 serial dilutions.
Antibodies were incubated on the cells at 37 C with 5% CO2 for 48 hours. Then 100 pl CellTiter-Glo reagent was added to each well and mixed well. After incubation of another 10 minutes at room temperature luminescence signal was read in a Spectra Max M5 plate reader.
Figure 37 shows the analysis of inhibition of cell proliferation (Cell TiterGlo Assay) of three different human tumor cells (DLD-1, NCI H460 and MDA-MB-231) upon treatment with different, cross-linked DR5 antibodies at a concentration of 7 nM. Figure 38 shows apoptosis induction measured by Caspase 8 activation in three human tumor cell lines (DLD-1, NCI H460 and MDA-MB-231) after treatment with cross-linked DRS antibodies at a concentration of 7 nM.
All tested DRS antibodies induce high caspase 8 activation in all tested tumor cell lines. DRS
antibodies were able to decrease the viability of all tested cell lines with varying potencies.
Example 28: Evaluation of chemical stability of DR5 antibodies derived by phage display and immunization Generation of stressed DR5 antibody samples To test the chemical stability of DRS antibodies, stressed samples were generated and functionally characterized with regard to DRS binding. High-pH stress induces ¨ among others ¨
deamidation of reactive Asn hotspots, whereas at pH 6.0 e.g. succinimide formation from reactive Asn and Asp residues may be induced. For high-pH stress, samples were transferred in 20 mM Na-phosphate, pH 8.0 and incubated for 5 days at 40 C. For low-pH
stress, samples were transferred into in 20 mM His/HisCl, 140 mM NaC1, pH 6.0 and incubated for 3 weeks at 40 C.
A control sample was kept at -80 C.
Determination of the relative active concentration of stressed DR5 antibody samples Around 5000 resonance units (RU) of the capturing system (20 p g/ml goat anti rabbit;
ordering code JIR111-005-046; Jackson Immuno Research) were coupled on a CMS
chip (GE
Healthcare BR-1005-30) at pH 5.0 by using an amine coupling kit supplied by the GE
Healthcare. The sample and system buffer was PBS-T (10 mM phosphate buffered saline including 0.05% Tween20) pH 7.4. The temperature of the flow cell was set to 25 C and of the sample block to 12 C. Before capturing, the flow cell was primed with running buffer twice.
The bispecific antibody was captured by injecting a 50 nM solution for 60 sec at a flow of 10 t1/min. Association was measured by injection of human DRS in solution for 90 sec at a flow of 30 t1/min at a concentration of 200 nM. The dissociation phase was monitored for up to 90 sec and triggered by switching from the sample solution to running buffer. The surface was regenerated by 60 sec washing with a 0.85% H3PO4 (phosphoric acid) solution at a flow rate of
-176-30 t1/min. Bulk refractive index differences were corrected by subtracting the response obtained from a blank surface.
The relative active concentration of the stressed antibody is the ratio calculated from the capture level and binding level (RU binding divided by RU capture, in comparison to the unstressed reference sample).
Figure 39 shows an exemplary response curve used for the determination of the relative active concentration of stressed DRS antibody samples. Figure 40 and 41 show relative active concentrations of original and stressed DRS antibodies derived from immunization and from phage display. Besides antibodies DRSTAA-0013 and DRSTAA-0010 that showed slightly deminished relative active concentrations after stress at pH6 and pH8, respectively, all other DRS antibodies did not show any relevant reduction of relative active concentrazion after pH
stress.
Example 29: Functional characterization of DR5-binder derived by immunization and phage display in 2+2 bispecific format in co-culture assays In order to evaluate whether the novel DRS binders derived by immunization can be used for the generation of bispecific antibodies for the targeted induction of apoptosis of tumor cells by hyper-cross-linking of DRS, a set of DRS antibodies were converted into 2+2 bispecific molecules (as depicted in fig 18 A) in combination with the FAP antibody 28H1.
Bispecific constructs were tested for their apoptosis inducing activity in a co-culture assay.
DLD-1 or H460 tumor cells (10,000 cells/well) are seeded to 96-well plates in co-culture with 3T3 cells or with 3T3 cells transfected to express murine FAP 2,500 cells/well) in a total volume of 150 1 to allow for triplicate samples for each treatment. After 24h, cells are treated with 50 1 antibodies (4X concentration) for 24h (untreated control: 50 1 medium).
Cell Death Detection ELISA: (Roche Applied Science Cat. No. 11774 425 001):
Procedure is followed exactly as stated in manufacturer's protocol. Vmax values are extrapolated from the measurement of abs at 405nm (minus reference wavelength value at 490nm) every minute for 10 minutes. An average of triplicate background values (lysis buffer alone) are subtracted from all samples. Data is expressed as fold increase over apoptosis in untreated samples.
Figure 43 shows induction of apoptosis in DLD-1 and H460 tumor cell lines by 2+2 bispecific constructs in co-culture assays as detected by DNA fragmentation.
While a bispecific construct containing Drozitumab as DRS-binding component already induces apoptosis in the absence of FAP, all constructs containing new DRS binders derived by immunization only induce apoptosis in the presence of FAP. Constructs with newly developed DRS
binders, such as
-177-0011-28H1 and 0016-28H1, are able to induce apoptosis to a higher extent especially at low concentrations as compared to the Drozitumab containing bispecific construct.
Example 30: Characterization of cellular binding of DR5-binder derived by immunization and phage display in 2+2 bispecific format Cells were stained (5x104/50 1) with 24 tg/m1 of each DRS-FAP construct or Drozitumab in staining buffer (PBS + 5% FCS) for 1 h on ice. After washing twice the secondary antibody goat anti-human IgG-AF488 (Invitrogen #A11013) was added at 10 tg/m1 and cells were again incubated 1 h on ice protected from direct light. After two more washing steps cells were measured at FACS Canto.
All tested constructs show binding to MDA-MB-231 with varying intensities (see Figure 44).
Example 31: Humanization of the VH and VL domains of DR5 antibodies derived by immunization The rabbit DRS-binding antibody DRSTAA-0011 was humanized using frameworks identical to human germline sequences. The human germline sequences hVH_3_64 (GenBank accession No.
M99682) and hVH3_16 (GenBank accession No P01767) were the 2 acceptors for the VH
humanized variants and the human germline sequences hVK1_93 (Accession No.
P04431) and hVK1_5 (GeneBank accession No. P01602) were the acceptors for VL humanization.
Eight humanized DRS antibodies comprising a heavy chain variable region construct selected from SEQ ID NOs. 23 and 26, and a light chain variable region construct selected from SEQ ID NOs 24, 29, 30, 31, and 32 were obtained and further characterized (Examples 27 to 29).
Table 24: Humanized DRS binders derived by immunization Variant HC SEQ ID LC SEQ ID
variant NO. variant NO.
-178-Example 32: Functional characterization of humanized DR5-binder derived by immunization Humanized DR5 antibodies were functionally characterized by evaluating apoptosis as measured by Cell Death Detection ELISA (CDDE, Roche #11 774 425 001) upon treatment with humanized anti-DRS antibodies in the presence or absence of a cross-linking antibody.
Preparation of cells: the adherent MDA-MB-231 cell line (human breast adenocarcinoma) was grown in DMEM medium (PAN) supplemented with 10% fetal calf serum (Gibco) and 2mM L-glutamine (PAA), and normally split twice per week 1:10. For the assay, cells were washed with PBS, detached from the flask with Accutase (PAA), seeded in 96-well flat-bottomed microtiter plates (Costar) at a density of 1x104 cells/well (CDDE) in 50p1 and incubated overnight at 37 C, 5% CO2.
Induction of apoptosis by humanized anti-DR5 antibodies: samples (humanized anti-DRS IgGs, DRS-TAA-#) were added to the cells at different concentrations alone or together with the cross-linking antibody (goat anti-human IgG, Sigma #I2136) in a 1:1 ratio in 50p1 in PBS, to induce crosslinking of the DRS receptors leading to apoptosis. Cells were incubated at 37 C for 24 hs (CDDE).
Cell Death Detection Elisa (CDDE): the immunoassay was performed according to the manufacturer's instructions (Roche). Briefly, supernatants were carefully aspirated and cells lysed with 200p1/well lyse buffer for 30 minutes at RT. A master mix consisting of anti-histone and anti-DNA antibodies was prepared according to the manufacturer's instructions and mixed with the 1:4-diluted lysates on 96-well streptavidin-coated flat-bottomed microtiter plates (Roche). After a 2-hour incubation at RT, wells were washed, ABTS substrate added and incubated at RT until color development sufficient for photometric analysis (10-30 min).
Absorbance was read at 405nm with a Tecan Spectra Rainbow Reader. Apoptosis signals were normalized to the apoptosis of the chimeric anti-DRS antibody at a concentration of 2p g/ml.
Results are shown in Figure 45: A Humanized Variants of DR5TAA-0011 (DR5TAA-0066 ¨
DR5TAA-0075, black lines) induce apoptosis upon crosslinking with secondary antibody in a dose-dependent manner. Several humanized variants, such as DR5TAA-0067, DR5TAA-0071, DR5TAA-0074 and DR5TAA-0075, are able to induce apoptosis in a similar manner concerning maximum of induction and dose-dependency as compared to the chimeric variant (DR5TAA-0052, grey lines). B Humanized Variants of DR5TAA-0011 (DR5TAA-0066 ¨ DR5TAA-0075) induce no apoptosis if not crosslinked by a secondary antibody. Thus, also the humanized
-179-variants are suited to be used in bispecific formats to specifically induce apoptosis only in the presence of FAP.
The Fabs of selected humanized variants were were fused C-terminally to a human Fc region via a (G4S)4 connector. These molecules were transiently produced in cells, purified via ProteinA beads and tested in an apoptosis induction assay.
In figure 22 b) the results of the DNA fragmentation assay in MDA-MB-231 cells with these Fe-DRS
fusion molecules after cross-linking with secondary anti Fc IgG are summarized. All tested molecules are able to induce apoptosis of the target cell line, indicating that the chosen DRS binders are not N-terminally blocked which opens a wider range of formats that can be used with these binders.
Example 33: Characterization of binding affinities of humanized DR5-binder derived by immunization Humanized DRS-binder were characterization for their binding affinities by SPR
analysis.
BIAcore characterization: A BIAcore 3000 instrument (GE Healthcare) was used with a CMS
sensor mounted into the system. The sensor was preconditioned by a 1 min injection at 100 p1/min of 0.1 % SDS, 50 mM NaOH, 10 mM HC1 and 100 mM H3PO4. System buffer was mM HEPES pH 7.4, 150 mM NaC1, 0.05% TWEEN 20. The sample buffer was the system buffer supplemented with 1 mg/mL carboxymethyldextran (Sigma). An anti-human antibody capture system was established on the biosensor surface. 8000 relative response units of a goat anti-human Fcy fragment-specific polyclonal antibody (Jackson Laboratories) were immobilized according to the manufacturer's instructions using EDC/NHS chemistry. The sensor was deactivated using 1M ethanolamine.
20 nM of the respective antibody sample were captured for 1 min at a flow rate of 10 t1/min. As a reference, 20 nM polyclonal human normal IgG (Roche, Ident. 11717570) were captured on the reference flow cell 1 and subtractive signals were monitored.
In one embodiment, the 23.3 kDa analyte DRS was injected at 30 t1/min for 3 min association time in concentration series at 0 nM, 3.3 nM, 11 nM, 2 x 33 nM, 100 nM and 300 nM. The complex dissociation was monitored for 5 min.
The system was regenerated at 30 t1/min by a 1 min injection of 10 mM glycine buffer pH 1.5 followed by a two consecutive 1 min injections of 10 mM glycine buffer pH 1.7.
Kinetic parameters were evaluated using the Biacore Evaluation Software according to the manufacturer's instructions.
The association rate constant ka(1/Ms), the dissociation rate constant kd (1/s) and the dissociation constant KD were calculated according to a Langmuir model with RmAx global.
-180-Table 25: Kinetic affinities to monomeric human DRS of humanized variants of (DRSTAA-0067 to DRSTAA-0075) as compared to chimeric form of DRSTAA-0011 (DRS TAA-0052) Construct SEQ ID ka kd (1/s) KD (nM) No (1/Ms) VH/VL
DR5TAA-0067 23/24 2,6E05 5.2E-03 20 DR5TAA-0074 23/31 2.7E05 5.6E-03 21 DR5TAA-0071 26/24 3.0E05 4.6E-03 15 DR5TAA-0075 23/29 3.2E05 5.6E-03 17 DR5TAA-0052 90/92 3.5E05 2.4E-03 7 Kinetic binding properties of humanized variants were maintained within a factor of approx. 3 for KD and kd and within a factor of approx. 2 for ka as compared to the chimeric variant.
Example 34: Characterization of thermal and chemical stability of humanized DRS-binder derived by immunization Humanized DRS-binder were characterized for their thermal and chemical stability. Samples were prepared at a concentration of 1 mg/mL in 20 mM Histidine/Histidine chloride, 140 mM
NaCE pH 6.0, transferred into an optical 384-well plate by centrifugation through a 0.4 p m filter plate and covered with paraffin oil. The hydrodynamic radius is measured repeatedly by dynamic light scattering while the samples are heated with a rate of 0.05 C/min from 25 C to 80 C. The aggregation onset temperature is defined as the temperature at which the hydrodynamic radius starts to increase.
Table 26: Aggregation Onset temperatures of humanized variants of DRSTAA-0011 (DRSTAA-0066-DRSTAA-0075) as a measure for thermal stability of the antibodies.
Construct SEQ ID No Tagg [ C]
VH/VL
-181-Humanized variants of DR5TAA-0011 reveal a high thermal stability with aggregation temperatures of 68 C and higher.
Chemical Stability Generation of stressed DR5 antibody samples: To test the chemical stability of DRS
antibodies, stressed samples were generated and functionally characterized with regard to DRS
binding. High-pH stress induces ¨ among others ¨ deamidation of reactive Asn hotspots, whereas at pH 6.0 e.g. succinimide formation from reactive Asn and Asp residues may be induced. For high-pH stress, samples were transferred in 20 mM Na-phosphate, pH 8.0 and incubated for 5 days at 40 C. For low-pH stress, samples were transferred into in 20 mM
His/HisCl, 140 mM
NaC1, pH 6.0 and incubated for 3 weeks at 40 C. A control sample was kept at -80 C.
Analysis of stressed DR5 antibody samples: A BIAcore 3000 instrument (GE
Healthcare) was used with a CMS sensor mounted into the system. The sensor was preconditioned by a 1 min injection at 100 t1/min of 0.1 % SDS, 50 mM NaOH, 10 mM HC1 and 100 mM H3PO4.
System buffer was 10 mM HEPES pH 7.4, 150 mM NaC1, 0.05% TWEEN 20. The sample buffer was the system buffer supplemented with 1 mg/mL carboxymethyldextran (Sigma). An anti-human antibody capture system was established on the biosensor surface. 8000 relative response units of a goat anti-human Fcy fragment-specific polyclonal antibody (Jackson Laboratories) were immobilized according to the manufacturer's instructions using EDC/NHS
chemistry. The sensor was deactivated using 1M ethanolamine.
20 nM of the respective antibody sample were captured for 1 min at a flow rate of 10 t1/min. As a reference, 20 nM polyclonal human normal IgG (Roche, Ident. 11717570) were captured on the reference flow cell 1 and subtractive signals were monitored.
In another embodiment, the analyte was injected at 100 t1/min for 2 min association time at 0 nM and 500 nM. The complex dissociation was monitored for 5 min.
Table 27: Relative active concentrations of humanized variants of DR5TAA-0011 before and after stress test Construct SEQ II) Initial pH 6.0 2 weeks 40'C
pll 7.4 2 we 40'C
No VIVVL (defined) Relative active Relative active concentration as concentration as compared to the initial compared to the initial state state DR5TAA- 26/24 100% 103% 98%

DR5TAA- 23/24 100% 107% 107%
-182-DR5TAA- 23/31 100% 103% 100%

DR5TAA- 26/30 100% 104% 101%

DR5TAA- 26/31 100% 103% 101%

DR5TAA- 26/32 100% 105% 101%

DR5TAA- 23/30 100% 102% 100%

DR5TAA- 23/29 100% 102% 99%

The system was regenerated at 30 t1/min by a 1 min injection of 10 mM glycine buffer pH 1.5 followed by a two consecutive 1 min injections of 10 mM glycine buffer pH 1.7.
Kinetic parameters were evaluated using the Biacore Evaluation Software according to the manufacturer's instructions.
The antibody/antigen complex half-life was calculated in minutes according to the formula ln(2)/(60*kd). The Molar Ratio was calculated: MW (antibody) / MW (antigen) *BL (antigen)/
CL (antibody).
Data report points were recorded shortly before the end of the antibody injection (antibody capture level, CL) as well as shortly before the analyte (Binding Late, BL) injection. Capture Level (CL) and Binding Late (BL) response signal heights were used to characterize the antibody binding performance. The relative binding quotient was calculated BL/CL. A quotient was formed from the relative binding quotient of an antibody sample versus a non-stress impacted antibody sample (relative active binding).
Results are shown in Table 27. Humanized variants of DR5TAA-0011 were subjected to stress test. None of the humanized variants showed impaired binding to human DR5 as compared to the non-stressed initial material.
Example 35: Materials and Methods Unless otherwise mentioned the following materials and methods have been used in the experiments outlined above.
Recombinant DNA technologies All antibody and antigen expression vectors were generated using standard recombinant DNA technology as described in Sambrook, J. et al., Molecular cloning: A
laboratory manual;
-183-Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989.
Molecular biological reagents were used according the manufacturer's recommendations.
Genes or gene fragments were either amplified by polymerase chain reaction (PCR) or generated from synthetic oligonucleotides at Geneart AG (Regensburg, Germany) by automated gene synthesis. PCR-amplified or subcloned DNA fragments were confirmed by DNA sequencing (Synergene GmbH, Switzerland). Plasmid DNA was transformed into and amplified in suitable E.
coli host strains for preparation of transfection-grade plasmid DNA using standard Maxiprep kits (Qiagen). For production of the bispecific molecules HEK293 EBNA cells were transfected with plasmids encoding the respective genes using a standard polyethlenimine (PEI) based method. The used plasmid ratio of the three expression vectors was 1:1:1. Transfected cells were cultivated for 7 days before supernatants were harvested for purification.
Transfection HEK293 EBNA cells All (bispecific) antibodies and antigens (if not obtained from a commercial source) used herein were transiently produced in HEK 293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below.
HEK293-EBNA cells are cultivated in suspension serum free in CD CHO culture medium.
For the production in 500 ml shake flask 400 million HEK293- EBNA cells are seeded 24 hours before transfection. For transfection cells are centrifuged for 5 min by 210 x g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD
CHO medium to a final amount of 200 ug DNA. After addition of 540 n1 PEI
solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature.
Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37 C in an incubator with a 5 % CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7 % Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210 x g, the solution is sterile filtered (0.22 nm filter) and sodium azide in a final concentration of 0.01 % w/v is added, and kept at 4 C.After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 nm sterile filters and stored at 4 C until purification.
Purification: standard + ProtA beads The proteins were produced by transient expression in HEK293 EBNA cells. All bispecific molecules described here were purified in two steps using standard procedures, such as proteinA
affinity chromatography (Akta Explorer) and size exclusion chromatography (Superdex 200).
The supernatant was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to Mabselect Sure resin (GE Healthcare) packed in a Tricorn 5/50 column (GE Healthcare, column volume (cv) = 1 ml) equilibrated with buffer A (50 mM sodiumphosphate, pH 7.0, 250 mM
NaC1).
-184-Unbound protein was removed by washing with at least 5 column volumes (CV) of buffer A.
The protein of interest was eluted in a linear pH-gradient from 0-100% buffer B (50 mM
sodiumphosphate, pH 7.0, 1 M NaC1) over 12 CV. Finally, an additional step was included with CV of buffer B followed by an equilibration step of 5 CV buffer A. Fractions containing the protein of interest were pooled and the pH was gradually adjusted to pH 6.0 (using 2 M TRIS
pH 8.0). Samples were concentrated to 0.5-2 ml using ultra-concentrators (Vivaspin 15R
30.000 MWCO HY, Sartorius) and subsequently applied to a HiLoadTm 16/60 SuperdexTm 200 preparative grade column (GE Healthcare) equilibrated with 20 mM Histidine, pH
6.0, 140 mM
NaC1, 0.01% Tween-20. The aggregate content of eluted fractions was analyzed using a TSKgel SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, mM NaC1, 200mM L-Arginine Monohydrocloride, 0.02 % (w/v) NaN3, pH 6.7 at 25 C.

Fractions containing less than 2 % oligomers were pooled and concentrated to a final concentration of 1 - 1.5 mg/ml using ultra concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius). Purified proteins were frozen in liquid N2 and stored at -80 C.
For a fast and high throughput purification, supernatants were neutralized using 1/40th column of 2M Tris-HC1 pH8 and incubated with ProteinA Sepharose Fast Flow beads (GE
Healthcare Cat No. 17-5138-01)) for 19h at 4 C end over end. The supernatant/bead mixture was then passed over an empty, equilibrated PD-10 column (GE Healthcare Cat No. 17-0435-01) by gravity flow. The retained beads were washed twice with binding buffer (10 mM
Tris, 50 mM
glycine, 100 mM NaC1, pH 8.0) and the antibody eluted with a low pH step (10 mM Tris, 50 mM
glycine, 100 mM NaC1, pH2.5). Finally the eluted protein was neutralized by addition of 1/40th volume of 2M Tris-HC1 pH8Ø The protein concentration of purified antibodies was calculated from the measured absorbance at 280 nm and the molar extinction coefficient calculated from the amino acid sequence. The aggregate content of the antibody sample was analysed using a Zorbax GF-250 analytical size exclusion column (Agilent Cat No PSMO 845006) equilibrated in running buffer (200 mM sodium phosphate, 0.02% sodium azide pH 7.0) at 25 C.
FACS binding analysis All used target cell lines were analyzed for relative expression levels of tumor-related antigens and DRS death receptors before apoptosis assays were performed.
Number and viability of cells was determined. For this, adherently growing cells were detached with cell dissociation buffer (Gibco ¨ Invitrogen # 13151-014). Cells were harvested by centrifugation (4 min, 400 x g), washed with FACS buffer (PBS / 0.1 % BSA) and the cell number was adjusted to 1.111 X 106 cells / ml in FACS buffer. 180 n1 of this cell suspension was used per well of a 96 well round bottom plate, resulting in 2 x 105 cell per well. The cells were incubated for 30 min at 4 C with the first antibody in appropriate dilution.
Then the cells were harvested by centrifugation (4 min, 400 x g), supernatant was completely removed and cells were washed once with 150 n1 of FACS buffer. The cells were resuspended in 12 ittl diluted
-185-secondary antibody (in case unlabelled first antibody was used) or FACS buffer for 30 min at 4 C in the dark. After two washing steps with FACS buffer cells were resuspended in 200 ial of FACS buffer and analyzed in a HTS FACSCanto II (BD, Software FACS Diva).
Alternatively the cells could be fixed with of 200 ial of 2 % PFA (paraformaldehyde) in FACS
buffer for 20 min at 4 C and analyzed later. All assays were performed in triplicates.
Table 28: Antibodies and concentrations for FACS binding analysis Antibody Source Description Conc. Conc. in test fing / lug / mll mll 1. First antibodies anti hu DR5 (TRAIL R&D #MAB631 mu IgG1 , 0.5 5 - 10 R2) clone 71903 Drozitumab in house hu IgG1 3.8 10 4G8 in house hu IgG1 20.5 15 mouse anti-human FAP Calbiochem mu IgG1 1 10 #0P188 Drozitumab-scFab- in house 1.34 25 FAP
Drozitumab- 1.44 25 X-FAP_A
Drozitumab- 0.83 25 X-FAP_B
5E11_28H1_N-term 2.55 as indicated VHVL
5E11_28H1_N-term 1.12 as indicated 5E11_28H1_C-term 4.0 as indicated 2. Secondary antibodies:
PE-conjugated Jackson 1:20 AffiniPure F(ab')2 ImmunoResearch dilution Fragment goat anti- Lab # 109-116-human IgG Fcg 170 Fragment Specific FITC-conjugated Serotec # 1:20 F(ab')2 goat anti- STAR105F dilution mouse IgG Specific FITC-conjugated Jackson Immuno 1:20 AffiniPure F(ab')2 Research Lab # dilution Fragment goat anti- 109-096-098 human IgG Fcg Fragment Specific Biacore analysis (Surface Plasmon Resonance, SPR)
-186-Binding of the various anti-DR5 binders as IgG or in a bispecific format was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T100 at 25 C with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaC1, 3 mM
EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).
For the determination of the species cross-reactivity of the DR5 binders, biotinlyated DR5 from mouse, human and cynomolgus were directly coupled to different flow cells of a Streptavidin (SA) sensor chip with an immobilization level of approximately 300 RU each. The various DRS binders as IgGs were injected at a concentration of 500, 100 and 25 nM for 60 s with a flow rate of 30u1/min, followed by a dissociation phase of 90 s. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell, where no protein was immobilized.
In a second experiment, the specificity of the DRS binders was determined by immobilizing huDR4Fc, huDcRlFc, huDcR2Fc and rhuOPGFc to a CMS sensor chip by amine coupling. Immobilisation levels were between 100 and 400 RU. Each binder was passed over the different flow cells at a concentration of 500, 100 and 25 nM for 60 s with a flow rate of 30 1/min and the dissociation phase monitored for 90 s. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell, where no protein was immobilized.
Further, the avidity of the IgGs as well as the 2+2 formats was measured on a CMS chip with immobilized human and cynomolgus DRS ECD (immobilization levels were around 100 RU). Each construct or IgG was passed over the different flow cells at a concentration between 500-0.97 nM in 1:2 dilution steps for 90s at 30 1/min. The dissociation was analysed for 120 s.
Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell, where no protein was immobilized. Kinetic constants were calculated using the Biacore T100 Evaluation Software (vAA, Biacore AB, Uppsala/Sweden), to fit rate equations for 1:1 Langmuir binding by numerical integration either as kinetic analysis or steady state analysis.
Affinity was assessed using a capture format where either an anti-human Fab or anti-human Fc antibody (Biacore, Freiburg/Germany) was directly coupled on a CMS
chip at pH 5.0 using the standard amine coupling kit (Biacore, Freiburg/Germany). The immobilization level was about 8,000-10000 RU. DRS binders in an IgG or 2+2 format were captured at a concentration of 50 or 30 nM respectively for 60 s at a flow rate of 30 1/min. Injection of human or cynomolgus DRS in a concentration range from 500 - 0.975 nM
(cynomolgus DRS) or 1000 - 0.975 nM (huDR5) in 1:2 dilution steps for 120 s at 30 1/min was carried out for the 2+2 format. Affinity of the IgGs was only measured for huDR5 at a concentration from 2000 - 20 nM
in 1:3 dilution steps. Dissociation was evaluated over a period of 120 s. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell, where no protein was immobilized. Kinetic constants were calculated using the Biacore T100
-187-Evaluation Software (vAA, Biacore AB, Uppsala/Sweden), to fit rate equations for 1:1 Langmuir binding by numerical integration either as kinetic analysis or steady state analysis.
Epitope binning was measured in two different formats, either with a classical sandwich assay or with a tandem approach. In the classical sandwich assay, each DR5 binders is directly immobilized by amine coupling to one flow cell on the CMS chip surface with a target immobilization level of around 500 RU. Subsequently, huDR5 is passed over each flow cell at a concentraion of 500 nM for 60 s (flow rate 30 1/min) followed by an injection of another DRS
binder at a concentration of 30 nM for 60 s (flow rate 30 [d/min). The dissociation is monitored over a period of 60 s with the same flow rate. Injection of the same DRS
binder than the immobilized one is used as a control as this should not lead to a response increase if all DRS is bound by the immobilized binder.
In the tandem approach, huDR5ECD was immobilized on a CMS chip with a final response of 250 RU. The first DRS binder was then passed over the flow cell at a concentration of 20 nM for 90 s, followed by the injection of a second DRS binder for 90 s.
The dissociation was monitored over a period of 90 s. The flow rate was 30 1/min for all steps. If the two binders recognize a different epitope, one could observe an increase in the response units. Injection of the same DRS binder was used as a control to confirm that all DRS molecules were saturated by the first injection and no additional binding to the same epitope can occur.
To further determine if any of the binders are ligand blocking, rhuTRAIL
(Peprotech Cat No. 310-04) was immobilized on a CMS chip by amine coupling with an immobilization level of 2000 RU. huDR5 Fc or huDR5 ECD were complexed with each DRS binder to be tested (100 nM DRS with 500 nM IgG) and the complex passed over the flow cell for 90 s with a flow rate of 50 it1/min. The dissociation was assessed over a period of 120 s. In addition, a classical sandwich assay was used, where huDR5Fc or huDR5 ECD was injected first at 100 nM followed by an injection of each DRS binder at 500 nM. Contact times were 60 s for DRS
and 90 s for the IgGs with a flow rate of 30 1/min. The dissociation step was carried out for 90s. Binders which could bind to TRAIL, in addition to DRS, were considered to be non-ligand blocking whereas binders which did not show any additional bind as ligand blocking.
Simultaneous binding of the various DRS binders in a 2+2 format was confirmed on a SA
chip containing immobilized huDR5Fc biotin (immobilisation level around 1000 RU). In a first step, the 2+2 construct was injected for 90 s followed by an injection of either human or murine FAP at a concentration of 500 or 100 nM for 90 s. The dissociation was monitored for 60 s. The flow rate for all steps was 30 it1/min. Simultaneous binding was considered to be true if an additional increase in response units was observed upon injection of human or murine FAP.
Epitope binning was measured in two different formats either a classical sandwich assay or with a tandem approach. In the classical sandwich assay each DRS binders gets directly immobilized by amine coupling to one flow cell on the CMS chip surface with a target immobilization level of around 500 RU. Then, huDR5 is passed over each flow cell at a conc. of
-188-500 nM for 60 s (flow rate 30 ul/min) followed by an injection of another DR5 binder at a concentration of 30 nM for 60 s (flow rate 30 ul/min). The dissociation is monitored over a period of 60 s with the same flow rate. Injection of the same DRS binder than the immobilized one is used as a control as this should not lead to a response increase if all DRS is bound by the immobilized binder.
In the tandem approach huDR5ECD was immobilized on a CMS chip with a final response of 250 RU. The first DRS binder is then passed over the flow cell at a concentration of 20 nM for 90 s, followed by the injection of a second DRS binder for 90 s. The dissociation was monitored over a period of 90 s. The flow rate was 30 ul/min for all steps. If the two binders recognize a different epitope, one could observe an increase in the response units.
Injection of the same DRS
binder was used as a control to confirm that all DRS molecules were saturated by the first injection and no additional binding to the same epitope can occur.
To further determine if any of the binders are ligand blocking rhuTRAIL
(Peprotech Cat No. 310-04) was immobilized on a CMS chip by amine coupling with an immobilisation level of 2000 RU. huDR5 Fc or huDR5 ECD were complexed with each DRS binder to be tested (100 nM DRS with 500 nM IgG) and the compley was passed over the flow cell for 90 s with a flow rate of 50 ul/min. The dissociation was assessed over a period of 120 s. In addition, a classical sandwich assay was used, where huDR5Fc or huDR5 ECD was injected first at 100 nM followed by an injection of each DRS binder at 500 nM. Contact times were 60 s for DRS
and 90 s for the IgGs with a flow rate of 30 ul/min. The dissociation step was carried out for 90s. Binders which could bind in addition of DRS to TRAIL were considered to be non-ligand blocking whereas binders which did not show any additional bind as ligand blocking.
Simultaneous binding of the various DRS binders in a 2+2 format was confirmed on a SA
chip containing immobilized huDR5Fc biotin (immobilisation level around 1000 RU). In a first step, the 2+2 construct was injected for 90 s followed by an injection of either human or murine FAP at a concentration of 500 or 100 nM for 90 s. The dissociation was monitored for 60 s. The flow rate for all steps was 30 t1/min. Simultaneous binding was considered to be true if an additional increase in response units was observed upon injection of human or murine FAP.
DNA fragmentation ELISA
For determination of induced apoptosis the Cell Death Detection ELISA PLUS kit from Roche was used. In short, 104 FAP expressing GM05389 cells per well of a 96-well plate (after detaching, and determination of cell number and viability) were seeded in 200 n1 appropriate medium and were incubated over night at 37 C in a 5 % CO2 atmosphere. The next day the medium was replaced by 100 ittl of fresh medium containing the apoptosis inducing antibodies, control antibodies and other controls in appropriate concentrations:
-189-The bispecific antibodies and IgGs were used in a final concentration of 0.7 and 7 nM or as indicated; cross-linking antibodies were used at the same molarity as the primary antibodies.
After addition of the antibodies104 apoptosis sensitive tumour cells were added per well.
The cells were incubated for 24 hrs at 37 C, 5 % CO2 to allow induction of apoptosis. The cells were harvested by centrifugation (10 min, 200 x g) and incubated for 1 h at room temperature in 200 n1 of lysis buffer (supplied by the kit). Intact DNA and the lysed cells were sedimented by centrifugation (10 min, 200 x g) and 20 n1 of the supernatant containing the fragmented DNA was analyzed according to the manufacturer's recommendations for induction of apoptosis.
Inhibition of proliferation (CellTiterGlo) For Cell Viability assays 4,000 tumor cells /well (in 75 1 volume) were seeded in black, clear bottom 96 well plates (BD Falcon cat#BD353220) and incubated overnight at 37 C in a humidified, 5% CO2 atmosphere. Then 25 1 4x DRS binder plus/minus rabbit-Fc (equal nano molar of DRS binders and Fc) with 6 concentrations of 2.5 x serial dilutions were added. After incubation at 37 C with 5% CO2 for 48 hours 100 pi CellTiter-Glo reagent was added to each well and mixed well. After incubation for another 10 minutes at room temperature results cell viability was determined with a Spectra Max M5 plate reader under luminescence settings.
Induction of Caspase 8 (Caspase Glo) For Caspase 8 activation assays 10,000 cancer cells/well in 75 n1 were seeded in opaque white 96 well plates (BD Falcon cat#BD353296) and incubated at 37 C in a humidified incubator with 5% CO2 overnight. Then 25u1 4x DRS binders plus/minus anti-Fc were added (equimolar ratios of DRS binders and anti Fc) with 6 concentrations of 2.5x serial dilutions.
After 3 hours incubation at 37 C with 5% CO2 100 pi caspase 8 substrate in lysis buffer was added to each well. After additional incubation for 30 minutes at 37 C the results were red in a Spectra Max M5 plate reader under luminescence settings.
FRET assays The binding of bispecfic molecules on cells was determined using a time-resolved fluorescence resonance energy transfer (TR-FRET) assay (called TagLite). Hek EBNA cells were grown to 60-80% confluency and transfected with plasmid DNA encoding for DRS ECD
fused to a SNAP Tag and the MalE TM. Briefly, 2 mg DNA was mixed with 4 ml OptiMEM
medium and 30 1 Lipofectamine 2000 (Invitrogen Cat No. 11668-019). The mixture was incubated for 20 min at RT. In the meantime, the adherent Hek EBNA cells grown in a T75 flask were washed with 5 ml D-PBS prior to adding the transfection mixture and culture medium (6 ml) (DMEM, 10% FCS, glutamax, Non-essential amino acids). The cells were then incubated overnight in a humidified incubator (5% CO2) at 37 C. Cells were washed with 5 ml D-PBS, followed by the addition of a mixture of 5 ml TagLite buffer (Cisbio Cat No.) containing 100 nM
-190-SNAP-Lumi4-Tb (Cisbio Cat No.). This resulted in attachment of the fluorescent dye to the SNAP Tag fused to the DR5. After an incubation time of 1 h at 37 C, the cells were washed with TagLite buffer to remove unbound dye. Subsequently, the labelling efficiency was checked by measuring the fluorescent signal at 620 nm (excitation 343 nm) of 10000 cells in a 384 well format (Reader, Victor, Perkin Elmer). Cells were then frozen in culture medium substituted with 10% DMSO and stored at -80 C.
To carry out a binding assay, pre-labeled cells were thawed, washed and 1000 cells per well mixed with 5 IA construct at a final concentration ranging from 50 -0.097 nM (1:2 dilution steps) and 5 t1 anti-huFc-d2 labeled (final concentration 150 nM). The fluorescent signal was measured at 620 nm for the fluorescent donor (Terbium) and at 665nm for the fluorescent acceptor dye after Oh, lh and 3h incubation at RT. The ratio of 665/620*1000 was calculated, and the reference (cells with 150 nM anti-huFc-d2) was subtracted. For KD
determination the results were analysed in Graph Pad Prism with one site fit-specific binding.
Determination of the thermal stability by Dynamic Light Scattering (DLS) Thermal stability of the protein is monitored by Dynamic Light Scattering (DLS). 30 p g of filtered protein sample with a protein concentration of 1 mg/ml is applied in duplicate to a Dynapro plate reader (Wyatt Technology Corporation; USA). The temperature is ramped from to75 C at 0.05 C/min, with the radius and total scattering intensity being collected.
Table 29: Names and Aliases of DRS Clones and Bispecific Constructs DR5-FAP 0R5 Clone FAP Clone Format Complete Scetch of bispecific (SEQ ID (SEQ ID antibody Format construct VH/VL) VH/VL) sequence DR5TAA-0061 DR5TAA-0011 4B9 1 + 1 SEQ ID NOs See Fig. 25 D
(SEQ ID (SEQ ID 280,281,282,283 NO.:88/89) NO.:39/40) DR5TAA-0030 5E11 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 131,132,124 NO.:7/8) NO.:15/16) DR5TAA-0032 DR5TAA-0005 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 284,285,286 NO.:41/46) NO.:15/16) DR5TAA-0033 DR5TAA-0011 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 287,288,289 NO.:88/89) NO.:15/16) DR5TAA-0034 DR5TAA-0013 28H1 2 + 2 SEQ ID NOs See Fig. 28A
(SEQ ID (SEQ ID 290,291,292 NO.:68/71) NO.:15/16) DR5TAA-0035 DR5TAA-0016 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 293,294,295 NO.:82/85) NO.:15/16) DR5TAA-0036 DR5TAA-0019 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 296,297,298 NO.:74/78) NO.:15/16)
-191-DR5TAA-0037 22E9 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 124,125,126 NO.:100/101) NO.:15/16) DR5TAA-0038 21H3 28H1 2 + 2 SEQ ID NOs See Fig. 28A
(SEQ ID (SEQ ID 127,128,124 -NO.:102/103) ., NO.:15/16) DR5TAA-0039 20F2 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID _ (SEQ ID 129,130,124 NO.:106/107) NO.:15/16) DR5TAA-0055 5E11 . 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 133, 132, 124 NO.:7/8) NO.:15/16) DR5TAA-0057 5E11 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 134,132,124 NO.:7/8) NO.:15/16) DR5TAA-0058 5E11 4B9 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 262,263,132 NO.:7/8) . NO.:39/40) DR5TAA-0077 5E11 28H1 2 + 2 SEQ ID NOs See Fig. 28 C
(SEQ ID (SEQ ID 135,136,137 NO.:7/8) - NO.:15/16) DR5TAA-0078 5E11 28H1 2 + 2 SEQ ID NOs 138, See Fig.

(SEQ ID (SEQ ID 139,137 NO.:7/8) NO.:15/16) , DR5TAA-0081 5E11 28H1 2 + 2 SEQ ID NOs See Fig. 28 F
(SEQ ID (SEQ ID 278,132,279 NO.:7/8) - NO.:15/16) DR5TAA-0117 DR5TAA-0067 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 299,300,301 NO.:23/24) : NO.:15/16) DR5TAA-0118 DR5TAA-0071 - 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID (SEQ ID 302,303,281,282, NO.:26/24) NO.:15/16) 283 , DR5TAA-0119 DR5TAA-0075 28H1 2 + 2 SEQ ID NOs See Fig. 28 A
(SEQ ID - (SEQ ID 305,306,307 NO.:23/29) NO.:15/16) DR5 Antibody DR5 Antibody Clone Name Clone Alias DR5TAA-0005 õ0005" or õ039"
DR5TAA-0006 õ0006" or õ058"
DR5TAA-0010 õ0010" or õ481"
DR5TAA-0013 "0013" or "298"
DR5TAA-0019 "0019" or "461"
DR5TAA-0016 "0016" or "422"
DR5TAA-0011 "0011" or "174"
Example 36: In vivo antitumor efficacy of DR5-FAP bispecific antibody comprising newly isolated DR5 binder 5E11 in combination with 28111 FAP CrossFab
-192-The in vivo antitumor efficacy of the bispecific antibody DR5-FAP (VH SEQ ID
NO. :7, VL
SEQ ID NO.: 8) was demonstrated in cell and fragment based patient derived (PDX) models of various tumor origin (e.g. CRC and pancreatic cancer) transplanted on nude mice. As example data are shown for the CRC xenograft model DLD-1 (cell line based, co-injection model) and Co5896 (fragment based).
Test agents The bispecific antibody DR5-FAP (DR5 binder: VH SEQ ID NO.:7, VL SEQ ID NO.:
8, FAP
binder: VH SEQ ID NO.:15, VL SEQ ID NO.: 16) was provided as stock solution from Roche, Penzberg, Germany. Antibody buffer included histidine. Antibody solution was diluted appropriately in buffer from stock prior injections.
Cell lines and culture conditions DLD-1 human CRC cells were originally obtained from ATCC. The tumor cell line was routinely cultured in DMEM high glucose medium with 1.0mM Sodium pyruvate supplemented with 10% fetal bovine serum, 2.0mM L-glutamine, 10mM HEPES at 37 C in a water-saturated atmosphere at 5% CO2. Culture passage was performed with trypsin / EDTA lx splitting every third day. Additionally murine fibroblasts NIH3T3 were purchased from ATCC and cultured in DMEM high glucose with 1.0mM Sodium pyruvate, FCS 10% and L-Glutamine 2.0mM.
Patient-derived xenograft model (PDX) The CRC tumor xenograft Co5896 was originally obtained from patients and passaged approximately three to five times until establishment of stable growth patterns. For the subsequent in vivo studies Co5896 tumor fragments were obtained from xenografts in serial passage in nude mice. After removal from donor mice, tumors were cut into fragments (4-5 mm diameter) and placed in PBS until subcutaneous implantation. Mice under isofluorane anesthesia received unilateral, subcutaneous tumor implants in the flank.
Animals Nude mice were purchased from breeder (e.g. Charles River, Sulzfeld, Germany) and maintained under specific-pathogen-free condition with daily cycles of 12 h light /12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). Experimental study protocol was reviewed and approved by local government. After arrival animals were maintained in the quarantine part of the animal facility for one week to get accustomed to new environment and for observation. Continuous health monitoring was carried out on regular basis.
Diet food (Provimi Kliba 3337) and water (acidified pH 2.5-3) were provided ad libitum.
Monitoring
-193-Animals were controlled daily for clinical symptoms and detection of adverse effects. For monitoring throughout the experiment body weight of animals was documented.
Treatment of animals Animal treatment started after animal randomisation after cell or fragment transplantation when median tumor size was about 100-200mm3. Antibody was administered as single agent at 10 or 30mg/kg i.v. once or twice weekly for several weeks depending on the model.
The corresponding vehicle was administered on the same days.
Antibody efficacy DLD-1 CRC co-injection cell line based xenograft model DLD-1 CRC xenograft bearing mice were treated with bispecific antibody DR5-FAP
(DRS
binder: VH SEQ ID NO.:7, VL SEQ ID NO.: 8, FAP binder: VH SEQ ID NO.:15, VL
SEQ ID
NO.: 16) from study day 9 to 20 at dosages of 10 and 1.0mg/kg for 4 times. As a result, treatment with bispecific antibody DR5-FAP (DR5 binder: VH SEQ ID NO.:7, VL
SEQ ID NO.:
8, FAP binder: VH SEQ ID NO.:15, VL SEQ ID NO.: 16) showed dose-related significant anti-tumor efficacy with strong anti-tumor efficacy against s.c. DLD-1 xenografts.
The Tumor Growth Inhibition (TGI) was calculated at 89% (10mg/kg) and 79% (1.0mg/kg), respectively. In contrast, after treatment with DRS Fc mutant antibody drozitumab PG, LALA
(10mg/kg, once weekly) no anti-tumor efficacy was noticed (see Figure 48). Similar results were obtained with high FAP content (Co-injection study with DLD-1 / NIH3T3 fibroblasts; Ratio 80/20, Figure 48) and low FAP content (Co-injection study with DLD-1 / MRCS fibroblasts; Ratio 30/70, data not shown).
Co5896 CRC fragment based patient derived xenograft model (PDX) Co5896 CRC xenograft bearing mice were treated with bispecific antibody DRS-FAP (DRS
binder: VH SEQ ID NO.:7, VL SEQ ID NO.: 8, FAP binder: VH SEQ ID NO.:15, VL
SEQ ID
NO.: 16) from study day 18 to 34 at dose of 30mg/kg for 6 times. As a result, treatment with bispecific antibody DRS-FAP (DRS binder: VH SEQ ID NO.:7, VL SEQ ID NO.: 8, FAP binder:
VH SEQ ID NO.:15, VL SEQ ID NO.: 16) showed significant anti-tumor efficacy with strong anti-tumor efficacy against s.c. Co5896 patient-derived xenografts. The Tumor Growth Inhibition (TGI) was calculated at 76% (see Figure 49). Similar results were obtained in other CRC cell models (data not shown).
Example 36: FAP prevalence in human tumors The prevalence of FAP in human tumors was evaluated by IHC to get an understanding on possible clinical use of bispecific DRS-FAP antibody.
-194-Rat anti-human Seprase antibody (IgG2a, clone D8) from Vitatex (MABS1001) was used to immunostain 2,5 FFPET sections from various tumour indications on the Ventana Benchmark XT. Sections were subjected to standard CC1 treatment followed by antibody incubation for 60 at 37 C at a concentration of 5 ug/mL in Dako antibody diluent (S3022) and positive staining was detected using the Ultraview DAB detection system (Ventana #760-4456).
Matched isotype antibody from Abcam (ab18450) was used as the negative control.
FAP+ stromal infiltrate was present in human tumors of different indications including head and neck squamous cell carcinoma (HNSCC), breast cancer, colorectal cancer (CRC), pancreatic cancer (PAC), gastric cancer, non-small-cell lung carcinoma (NSCLC) and Mesothelioma marking potentially interesting clinical indications for a bispecific DR5-FAP
antibody (Table 30).
Table 30: FAP prevalence in human tumors % cases with moderate to high Tumor Type n of samples investigated +
______________________ infiltrate Breast Cancer 77 105 triple negative BC 80 7 Gastric Cancer 68 28 Mesothelioma 60 10 Sequences 1. Amino acid sequences of phage display derived DR5 binders Description Amino acid sequence SEQ ID
NO.

(22E9)_VH RQAPGKGLEWVSAISGSGGSTYYADSVKGRFTISRDNS
KNTLYLQMNSLRAEDTAVYYCAKGVRISFDYWGQGT
LVTVSS

(22E9)_CDRH1
-195-Description mino acid sequence SEQ ID
NO. I

(22E9)_CDRH2 (22E9)_CDRH3 (22E9)_VL KPGQAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRL
EPEDFAVYYCQQGSNQPVTFGQGTKVEIK

(22E9)_CDRL1 (22E9)_CDRL2 (22E9)_CDRL3 (21H3)_VH RQAPGKGLEWVS AIS GS GGS TYYADS VKGRFTISRDNS
KNTLYLQMNSLRAEDMAVYYCAKGARVSFDYWGQG
TLVTVS S

(21H3)_CDRH1 (21H3)_CDRH2 (21H3)_CDRH3 (21H3)_VL KPGQAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRL
EPEDFAVYYCQQGSQPPITFGQGTKVEIK

(21H3)_CDRL1 (21H3)_CDRL2 (21H3)_CDRL3 (20F2)_VH RQAPGKGLEWVS AIS GS GGS TYYADS VKGRFTISRDNS
KNTLYLQMNSLRAEDTAVYYCAKGVRKGFDYWGQG
TLVTVS S

(20F2)_CDRH1 (20F2)_CDRH2 (20F2)_CDRH3 (20F2)_VL KPGQAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRL
EPEDFAVYYCQQGESPPPTFGQGTKVEIK

(20F2)_CDRL1
-196-Description 1mino acid sequence SEQ ID
NO. I

(20F2)_CDRL2 (20F2)_CDRL3 (5E11)_VH RQAPGKGLEWVSAISGSGGSTYYADSVKGRFTISRDNS
KNTLYLQMNSLRAEDTAVYYCAKGVRVSFDYWGQGT
LVTVSS

(5E11)_CDRH1 (5E11)_CDRH2 (5E11)_CDRH3 (5E11)_VL KPGQAPRLLIYGASSRATGIPDRFSGSGSGTDFTLTISRL
EPEDFAVYYCQQGTTHPITFGQGTKVEIK

(5E11)_CDRL1 (5E11)_CDRL2 (5E11)_CDRL3 (18F11)_VH RQAPGKGLEWVSAISGSGGSTYYADSVKGRFTISRDNS
KNTLYLQMNSLRAEDAAVYYCAKGVRKKFDYWGQG
TLVTVSS
DRS (18F11)_ SYAMS 1 DRS (18F11)_ AISGSGGSTYYADSVKG 2 DRS (18F11)_ GVRKKFDY 98 (18F11)_VL KPGQAPRLLIYGASSRATGIPDRFSGSGSGTDFTLTISRL
EPEDFAVYYCQQGQLPPITFGQGTKVEIK
DRS (18F11)_ RASQSVSSSYLA 4 DRS (18F11)_ GASSRAT 5 DRS (18F11)_ QQGQLPPIT 97 2. Amino acid sequences of non-functional phage display derived DR5 binders (CDRH1=SEQ ID NO.:1, CDRH2=SEQ ID NO.:2, CDRL1=SEQ ID NO.:4 and CDRL2=SEQ ID
NO.:5) Name mino icid sequence SEQ ID
-197-N() DR5 (IB12)_CDR H3 GFGYWYMDY 266 DR5 (1B12)_CDR L3 QQSGRRQT 267 DRS (19C12)_CDR H3 SIFYSTLDY 268 DRS (19C12)_CDR L3 QQQGWFQT 269 DRS (19D6)_CDR H3 VLGYASYDY 270 DRS (19D6)_CDR L3 QQQGWSTT 271 DRS (20E3)_CDR H3 GTRRGFDY 272 DRS (20E3)_CDR L3 QQGELTPVT 273 3. Amino acid sequences of FAP binders ______________________________________________________________________ Name Amino acid ,,ccitience SLQ 11) N() FAP(28H1)_VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSSHAMSWV 15 RQAPGKGLEWVSAIWASGEQYYADSVKGRFTISRDNS
KNTLYLQMNSLRAEDTAVYYCAKGWLGNFDYWGQG
TLVTVSS
FAP(28H1)_VL EIVLTQSPGTLSLSPGERATLSCRASQSVSRSYLAWYQQ 16 KPGQAPRLLIIGASTRATGIPDRFSGSGSGTDFTLTISRLE
PEDFAVYYCQQGQVIPPTFGQGTKVEIK
FAP(4B9)_VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWV 39 RQAPGKGLEWVSAIIGSGASTYYADSVKGRFTISRDNS
KNTLYLQMNSLRAEDTAVYYCAKGWFGGFNYWGQG
TLVTVSS
FAP(4B9)_VL EIVLTQSPGTLSLSPGERATLSCRASQSVTSSYLAWYQQ 40 KPGQAPRLLINVGSRRATGIPDRFSGSGSGTDFTLTISRL
EPEDFAVYYCQQGIMLPPTFGQGTKVEIK
FAP SHAMS
(28H1)_CDRH1 9 FAP AIWASGEQYYADSVKG
(28H1)_CDRH2 10 FAP GWLGNFDY
(28H1)_CDRH3 11 FAP RASQSVSRSYLA
(28H1)_CDRL1 12 FAP GASTRAT
(28H1)_CDRL2 13 FAP QQGQVIPPT
(28H1)_CDRL3 14 FAP SYAMS
(4B9)_CDRH1 33 FAP AIIGSGASTYYADSVKG
(4B9)_CDRH2 34 FAP GWFGGFNY
(4B9)_CDRH3 35
-198-Name Amino acid scqucncc SFQ II) NO
FAP RASQSVTSSYLA
(4B9)_CDRL1 36 FAP VGSRRAT
(4B9)_CDRL2 37 FAP QQGIMLPPT
(4B9)_CDRL3 38 4. Amino acid sequences of bispecific molecules comprising conventional DR5 binders Namc Amino acid sequence SI Q II) NO
Drozitumab_VH EV QLV QS GGG V ERPGGS LRLSCAASGPTFDDYAMSWV RQA 274 PGKGLEWVSGINWQGGSTGYADSVKGRVTISRDNAKNS LY
LQMNS LRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
SS
Drozitumab_VL SELTQDPAVSVALGQTVRITCSGDSLRSYYASWYQQKPGQA 110 PVLVIYGANNRPS GIPDRFS GS S SGNTAS LTITGAQAEDEADY
YCNS ADS SGNHVVFGGGTKLTVL
Drozitumab-3F2 EVQLVQSGGGVERPGGS LRLSCAASGFIFDDYAMSWVRQA 111 VHVL- scFv (HC) PGKGLEWVSGINWQGGSTGYADSVKGRVTISRDNAKNS LY
pETR6606 LQMNS LRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
S S AS TKGPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQS S GLY S LS SVVTVPS S SLGTQTYI

FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFF
LYSKLTVDKSRWQQGNVFSCS VMHEALHNHYTQKS LS LS P
GKSGGGGSGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGS
LRLSCAASGFTFS S YAM SWVRQAPGKC LEWVS AIS GS GGS T
YYADSVKGRFTISRDNSKNTLYLQMNS LRAEDTAVYYCAK
GWFGGFNYWGQGTLVTVS SGGGGSGGGGSGGGGSGGGGS
EIVLTQSPGTLS LYPGERATLS CRAS QS VTS SYLAWYQQKPG
QAPRLLINVGSRRATGIPDRFS GS GS GTDFILTIS RLEPEDFA
VYYCQQGIMLPPTFGCGTKVEIK
-199-Name Amino acid sequence SEQ 11) NO
Drozitumab-FAP EVQLVQSGGGVERPGGSLRLSCAASGFIFDDYAMSWVRQA 112 (4G8) PGKGLEWVSGINWQGGSTGYADSVKGRVTISRDNAKNSLY
VHVL-scFv (HC) LQMNSLRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
pETR7342 SSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI
CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS V
FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF
LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSP
GGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGF
TFSSYAMSWVRQAPGKCLEWVSAISGSGGSTYYADSVKGR

GQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTL
SLSPGERATLSCRASQSVSRSYLAWYQQKPGQAPRLLIIGAS
TRATGIPDRFSGSGSGTDFILTISRLEPEDFAVYYCQQGQVIP
PTFGCGTKVEIK
Drozitumab-FAP SELTQDPAVSVALGQTVRITCSGDSLRSYYASWYQQKPGQA 113 (4G8) PVLVIYGANNRPSGIPDRFSGSSSGNTASLTITGAQAEDEADY
VHVL-scFv (LC) YCNSADSSGNHVVFGGGTKLTVLGQPKAAPSVTLFPPSSEEL
pETR7344 QANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSK
QSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVA
PTECSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASG
FIFSSYAMSWVRQAPGKCLEWVSAISGSGGSTYYADSVKG
/ 1=1 \
RI-TISRDNSKNTLYLQMNSLRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGT

STRATGIPDRFSGSGSGTDFILTISRLEPEDFAVYYCQQGQVI
PPTFGCGTKVEIK
-200-Name Amino acid sequence SFQ
NO
Drozitumab-3F2 EVQLVQSGGGVERPGGSLRLSCAASGPTFDDYAMSWVRQA 114 VLCL_VHCH1- PGKGLEWVSGINWQGGSTGYADSVKGRVTISRDNAKNSLY
scFab (HC) LQMNSLRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
pETR7369 S S AS TKGPS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQS S GLY S LS S VVTVPS SSLGTQTYI
CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS V
FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
1¨r VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF
LYS KLTVDKS RWQQGNVFS C S VMHEALHNHYTQKS LS LS P
GKSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLYPGER
ATLS CRAS QS VTS S YLAWYQQKPGQAPRLLINVGSRRATGIP
DRFS GS GS GTDFTLTIS RLEPEDFAVYYCQQGIMLPPTFGQGT
KVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQS GNSQES VTEQDSKDS TYS LS S TLTLSKAD
YEKHKVYACEVTHQGLS S PVTKS FNRGEC S GGGS GGGS EGG
GS EGGGS EGGGS EGGGS GGGS GEVQLLES GGGLVQPGGS LR
LS CAAS GFTFS S YAMS WVRQAPGKGLEWVS AIS GS GGS TYY
ADS VKGRPTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGW
FGGFNYWGQGTLVTVS S AS TKGPS VFPLAPS SKS TS GGTAAL
GCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYS LS S
VVTVPS S S LGTQTYICNVNHKPS NTKVDKKVEPKS CD
Drozitumab-3F2 SELTQDPAVSVALGQTVRITCSGDSLRSYYASWYQQKPGQA 115 VLCL_VHCH1- PVLVIYGANNRPS GIPDRFS GS S SGNTAS LTITGAQAEDEADY
scFab (LC) YCNS ADS SGNHVVFGGGTKLTVLGQPKAAPS VTLFPPS SEEL
pETR7370 QANKATLVC LIS DFYPGAVTVAWKADS SPVKAGVETTTPSK
QS NNKYAAS S YLSLTPEQWKSHRS YSCQVTHEGS TVEKTVA
PTECSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLYPGE
RATLS CRAS QS VTS S YLAWYQQKPGQAPRLLINVGSRRATGI
PDRFS GS GS GTDFTLTIS RLEPEDFAVYYCQQGIMLPPTFGQG
, TKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQS GNSQES VTEQDSKDS TYS LS S TLTLSKAD
YEKHKVYACEVTHQGLS S PVTKS FNRGEC S GGGS GGGS EGG
GS EGGGS EGGGS EGGGS GGGS GEVQLLES GGGLVQPGGS LR
LS CAAS GFTFS S YAMS WVRQAPGKGLEWVS AIS GS GGS TYY
ADS VKGRPTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGW
FGGFNYWGQGTLVTVS S AS TKGPS VFPLAPS SKS TS GGTAAL
GCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYS LS S
VVTVPS S S LGTQTYICNVNHKPS NTKVDKKVEPKS CD
-201-N Lime Amino acid sequence SE() 11) NO
Drozitumab-FAP EVQLVQSGGGVERPGGSLRLSCAASGFIFDDYAMSWVRQA 116 (4G8) PGKGLEWVSGINWQGGSTGYADSVKGRVTISRDNAKNSLY
VLCL_VHCH1- LQMNSLRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
scFab (HC) S S AS TKGPS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS
pETR7371 WNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI
CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS V
FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF
LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSP
GKSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGER
ATLSCRASQSVSRSYLAWYQQKPGQAPRLLIIGASTRATGIP
DRFSGSGSGTDFTLTISRLEPEDFAVYYCQQGQVIPPTFGQGT
KVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQSGNSQES VTEQDSKDS TYS LS S TLTLSKAD
YEKHKVYACEVTHQGLSSPVTKSFNRGECSGGGSGGGSEGG
GSEGGGSEGGGSEGGGSGGGSGEVQLLESGGGLVQPGGSLR
LSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGSGGSTYY
ADS VKGRPTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGW
LGNFDYWGQGTLVTVS S AS TKGPS VFPLAPS SKS TSGGTAA
LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL
SSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCD
Drozitumab-FAP SELTQDPAVSVALGQTVRITCSGDSLRSYYASWYQQKPGQA 117 (4G8) PVLVIYGANNRPSGIPDRFSGSSSGNTASLTITGAQAEDEADY
VLCL_VHCH1- YCNS ADS SGNHVVFGGGTKLTVLGQPKAAPSVTLFPPS SEEL
scFab (LC) QANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSK
pETR7380 QSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVA
PTECSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGE
RATLSCRASQSVSRSYLAWYQQKPGQAPRLLIIGASTRATGI
PDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQGQVIPPTFGQG
TKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQSGNSQES VTEQDSKDS TYS LS S TLTLSKAD
YEKHKVYACEVTHQGLSSPVTKSFNRGECSGGGSGGGSEGG
GSEGGGSEGGGSEGGGSGGGSGEVQLLESGGGLVQPGGSLR
LSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGSGGSTYY
ADS VKGRPTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGW
LGNFDYWGQGTLVTVS S AS TKGPS VFPLAPS SKS TSGGTAA
LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL
SSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCD
-202-Name Amino acid sequence SE() 11) N
Drozitumab-FAP EVQLVQSGGGVERPGGS LRLSCAASGPTFDDYAMSWVRQA 118 (4G8) PGKGLEWVSGINWQGGSTGYADS VKGRVTISRDNAKNS LY
VHCL LQMNS LRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
2+2 S S AS TKGPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYI
CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS V
)¨( FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
led VEVHNAKTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTIS KAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFF
LYSKLTVDKSRWQQGNVFSCS VMHEALHNHYTQKS LS LS P
GKSGGGGSGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGS
LRLS CA AS GFTFS S YAM SWVRQAPGKGLEWVS AIS GS GGS T
YYADS VKGRFTISRDNS KNTLYLQMNS LRAEDTAVYYCAK
GWLGNFDYWGQGTLVTVS SAS VAAPS VFIFPPS DEQLKS GT
AS VVCLLNNFYPREAKVQWKVDNALQSGNSQES VTEQDSK
DS TYS LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSFNR
GEC
Drozitumab LC SELTQDPAVSVALGQTVRITCSGDSLRSYYASWYQQKPGQA 119 pETR7303 PVLVIYGANNRPS GIPDRFS GS S SGNTAS LTITGAQAEDEADY
YCNS ADS SGNHVVFGGGTKLTVLGQPKAAPS VTLFPPS SEEL
QANKATLVC LIS DFYPGAVTVAWKADS SPVKAGVETTTPSK
QS NNKYAAS SYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVA
PTECS
FAP (4G8) EIVLTQSPGTLS LS PGERATLS CRAS QS VS RS YLAWYQQKPG 120 _VLCH1 QAPRLLIIGASTRATGIPDRFS GS GS GTDFTLTIS RLEPEDFA V
YYCQQGQVIPPTFGQGTKVEIKS S AS TKGPS VFPLAPS S KS TS
GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS S
GLYS LS S VVTVPS S SLGTQTYICNVNHKPSNTKVDKKVEPKS
CD
Drozitumab -FAP EVQLVQSGGGVERPGGS LRLSCAASGPTFDDYAMSWVRQA 121 (4G8) PGKGLEWVSGINWQGGSTGYADS VKGRVTISRDNAKNS LY

2+2 S S AS TKGPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYI
CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS V
FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
)=r VEVHNAKTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTIS KAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFF
LYSKLTVDKSRWQQGNVFSCS VMHEALHNHYTQKS LS LS P
GKSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLS LS PGER
ATLS CRAS QS VS RS YLAWYQQKPGQAPRLLIIGAS TRATGIP
DRFSGSGSGTDFTLTISRLEPEDFAVYYCQQGQVIPPTFGQGT
KVEIKASTKGPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPV
TVSWNSGALTSGVHTFPAVLQS S GLYS LS S VVTVPS S SLGTQ
TYICNVNHKPSNTKVDKKVEPKSCD
-203-Name Aimno acid sequence 51:Q
11) N
FAP (4G8) EVQLLES GGGLVQPGGSLRLSCAASGFI FS S YAMSWVRQAP 122 _VHCL GKGLEWVS AIS GS GGS TYYADS VKGRFTIS RDNS KNTLYLQ
MNSLRAEDTAVYYCAKGWLGNFDYWGQGTLVTVS S AS VA
APS VFIFPPS DEQLKS GTASVVCLLNNFYPREAKVQWKVDN
ALQSGNSQESVTEQDSKDSTYS LS STLTLSKADYEKHKVYA
CEVTHQGLS SPVTKSFNRGEC
Drozitumab-FAP EVQLVQSGGGVERPGGSLRLSCAASGFI FDDYAMSWVRQA 123 (28H1) PGKGLEWVSGINWQGGSTGYADSVKGRVTISRDNAKNS LY
VHCL LQMNS LRAEDTAVYYCAKILGAGRGWYFDYWGKGTTVTV
pETR9551 S S AS TKGPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS
2+2 WNSGALTSGVHTFPAVLQS S GLY S LS SVVTVPS S SLGTQTYI
CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS V
FLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFF
LYSKLTVDKS RWQQGNVFS C S VMHEALHNHYTQKS LS LS P
GKSGGGGSGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGS
LRLSCAASGFTFS S HAM SWVRQAPGKGLEWVS AIWAS GEQ
YYADSVKGRFTISRDNSKNTLYLQMNS LRAEDTAVYYCAK
GWLGNFDYWGQGTLVTVS S AS VAAPS VFIFPPS DEQLKS GT
AS VVCLLNNFYPREAKVQWKVDNALQS GNS QES VTEQDS K
DS TYS LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSFNR
GEC
5. Amino acid sequences of bispecific molecules comprising phage display derived DR5 binders ______________________________________________________________________ Name Amino acid sequence SE() ID
N() FAP (28H1) EIVLTQS PGTLS LS PGERATLS CRA SQS VS RS YLAWYQQKPG 124 _VLCH1 QAPRLLIIGAS TRATGIPDRFSGSGSGTDF1 LTISRLEPEDFAV
pETR9537 YYCQQGQVIPPTFGQGTKVEIKS S AS TKGPS VFPLAPS S KS TS
GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS S
GLYS LS S VVTVPS S SLGTQTYICNVNHKPSNTKVDKKVEPKS
CD
-204-Name Amino acid ,,equence SEQ ID
N() DR5 (22E9)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAP 125 (28H 1) GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
VHCL MNS LRAEDTAVYYCAKGVRISFDYWGQGTLVTVS SAS TKG
pETR9711 PS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNS GALT
2+2 SGVHTFPAVLQS SGLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNS TYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
1=r PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
Tag GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKS RWQQ GNVFS CS VMHEALHNHYTQK S LS L S PGKS GGGG
SGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAAS
GI-TES SHAMS WVRQAPGKGLEWVSAIWASGEQYYADS VKG
RPTISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVS SAS VAAPS VFIFPPSDEQLKSGTAS VVCLLNN
FYPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS S TL
TLSKADYEKHKVYACEVTHQGLS SPVTKS FNRGEC
DR5 (22E9) LC EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPG 126 pETR9076 QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFA V
YYCQQGSNQPVTFGQGTKVEIKRTVAAPS VFIFPPSDEQLKS
GTAS VVCLLNNFYPREAKVQWKVDNALQ SGNSQES VTEQD
SKDS TYS LS S TLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC
DR5 (21 H3) -FAP EVQLLES GGGLVQPGGS LRLS CA AS GFTFS S YAMS WVRQAP 127 (28H 1) GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
VHCL MNS LRAEDMAVYYCAKGARVSFDYWGQGTLVTVS SAS TK
pETR10626 GPS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNS GAL
2+2 TSGVHTFPAVLQS SGLY S LS S VVTVPS S SLGTQTYICNVNHK
PS NTKVDKKVEPKS CDKTHTCPPCPAPELLGGP S VFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNS TYRVVS VLTVLHQDWLNGKEYKCKVSNKA
LPAPIEKTIS KAKGQPREPQVYTLPPS RDELTKNQ VS LTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKS RWQQ GNVFS CS VMHEALHNHYTQK S LS L S PGKS GGGG
SGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAAS
GI-TES SHAMS WVRQAPGKGLEWVSAIWASGEQYYADS VKG
RPTISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVS SAS VAAPS VFIFPPSDEQLKSGTAS VVCLLNN
FYPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS S TL
TLSKADYEKHKVYACEVTHQGLS SPVTKS FNRGEC
DR5 (21H3) LC EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPG 128 pETR9075 QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFA V
YYCQQGSQPPITFGQGTKVEIKRTVAAPS VFIFPPSDEQLKSG
TASVVCLLNNFYPREAKVQWKVDNALQS GNSQES VTEQDS
KDS TYS LS S TLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC
-205-Name Amin() acid ,,equence SEQ ID
N() DR5 (20F2)-FAP EVQLLES GGGLVQPGGS LRLS CA AS GFTFS S YAMS WVRQAP 129 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
VHCL MNS LRAEDTAVYYCAKGVRKGFDYWGQGTLVTVS S AS TK
pETR10135 GPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GAL
2+2 TSGVHTFPAVLQS SGLYS LS S VVTVPS S SLGTQTYICNVNHK
PS NTKVDKKVEPKS CDKTHTCPPCPAPELLGGPS VFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKA
1=r LPAPIEKTIS KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
Tag GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGKS GGGG
SGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAAS
GI-TES S HAMS WVRQAPGKGLEWVS AIWAS GEQYYADS VKG
RPTISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVS SAS VAAPS VFIFPPSDEQLKSGTAS VVCLLNN
FYPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS STL
TLSKADYEKHKVYACEVTHQGLS SPVTKS FNRGEC
DR5 (20E2) LC EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPG 130 pETR9061 QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFA V
YYCQQGESPPPTFGQGTKVEIKRTVAAPS VFIFPPSDEQLKSG
TASVVCLLNNFYPREAKVQWKVDNALQS GNSQES VTEQDS
KDSTYS LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC
DR5 (5E11 )-FAP EVQLLES GGGLVQPGGS LRLS CA AS GFTFS S YAMS WVRQAP 131 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
VHCL MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
pETR10334 PS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
2+2 SGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
1=r PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGKS GGGG
SGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAAS
GI-TES S HAMS WVRQAPGKGLEWVS AIWAS GEQYYADS VKG
RPTISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVS SAS VAAPS VFIFPPSDEQLKSGTAS VVCLLNN
FYPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS STL
TLSKADYEKHKVYACEVTHQGLS SPVTKS FNRGEC
DR5 (5E11) LC EIVLTQS PGTLS LS PGERATLS CRA SQS VS S SYLAWYQQKPG 132 pETR9044 QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFA V
YYCQQGTTHPITFGQGTKVEIKRTVAAPS VFIFPPSDEQLKSG
TASVVCLLNNFYPREAKVQWKVDNALQS GNSQES VTEQDS
KDSTYS LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC
-206-Name Amino acid sequence SEQ ID
N() DR5 (5E11)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 133 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRPTIS RDNS KNTLYLQ
VHCL MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
2+2 PS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
Removal of C- SGVHTFPAVLQS S GLY S LS SVVTVPS S SLGTQTYICNVNHKP
term. Lysine in Fc SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK
pETR11052 DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
1=r DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGGGGGS G
GGGSGGGGS GGGGSEVQLLESGGGLVQPGGS LRLSCAASGF
TES S HAMS WVRQAPGKGLEWVS AIVVAS GEQYYADSVKGRF
TISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDYW
GQGTLVTVS S AS VAAPS VFIFPPS DEQLKS GTA SVVCLLNNF
YPREAKVQWKVDNALQS GNS QES VTEQDS KDS TYS LS S TLT
LS KADYEKHKVYACEVTHQGLS SPVTKSFNRGEC
DR5(5E11)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 134 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRPTIS RDNS KNTLYLQ
VHCL MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
2+2 PS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
Removal of C- SGVHTFPAVLQS S GLY S LS SVVTVPS S SLGTQTYICNVNHKP
term. Lysine in Fc SNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPK

mut. TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
pETR11025 GAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGGGGGS G
GGGSGGGGS GGGGSEVQLLESGGGLVQPGGS LRLSCAASGF
TES S HAMS WVRQAPGKGLEWVS AIVVAS GEQYYADSVKGRF
TISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDYW
GQGTLVTVS S AS VAAPS VFIFPPS DEQLKS GTA SVVCLLNNF
YPREAKVQWKVDNALQS GNS QES VTEQDS KDS TYS LS S TLT
LS KADYEKHKVYACEVTHQGLS SPVTKSFNRGEC

DR5(5E11)-FAP QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFAV
(28H1) YYCQQGTTHPITFGQGTKVEIKS S AS TKGPS VFPLAPS S KS TS
pETR11827 GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS S
2+2 GLYS LS S VVTVPS S SLGTQTYICNVNHKPSNTKVDKKVEPKS
CDKTHTCPPCPAPELLGGPSVFLEPPKPKDTLMISRTPEVTCV
VVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR
VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ
PREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESN
GQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSC
-SVMHEALHNHYTQKS LS LS PGKS GGGGSGGGGSGGGGSGG
GGSEVQLLESGGGLVQPGGSLRLSCAASGFTFS S HAM SWVR
QAPGKGLEWVSAIVVASGEQYYADSVKGRFTIS RDNSKNTLY
LQMNSLRAEDTAVYYCAKGWLGNFDYWGQGTLVTVS S AS
TKGPSVFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVSWNS G
ALTSGVHTFPAVLQS S GLYS LS SVVTVPS S S LGTQTYICNVN
HKPSNTKVDKKVEPKS CD
-207-Name Amin() acid ,,equence SEQ ID
N() DR5 (5E11) EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 136 VHCL GKGLEWVSAISGSGGSTYYADSVKGRPTISRDNSKNTLYLQ
pETR11484 MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS VA
APS VFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDN
ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYA
CEVTHQGLSSPVTKSFNRGEC
FAP (28H1) EIVLTQSPGTLSLSPGERATLSCRASQSVSRSYLAWYQQKPG 137 VLCL QAPRLLIIGAS TRATGIPDRFSGSGSGTDFILTISRLEPEDFAV
pETR9366 YYCQQGQVIPPTFGQGTKVEIKRTVAAPS VFIFPPSDEQLKSG
TASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS
KDSTYS LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC

DR5(5E11)-FAP GKGLEWVSAISGSGGSTYYADSVKGRPTISRDNSKNTLYLQ
(28H1) MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS VA
pETR11828 APS VFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDN
2+2 ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYA
CEVTHQGLSSPVTKSFNRGECDKTHTCPPCPAPELLGGPSVF
LEPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKENVVYVDGV
1=r EVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK
VSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVS
LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFEL
YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG
GGGGSGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRL
SCAASGFTESSHAMSWVRQAPGKGLEWVSAIWASGEQYYA
DSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGWL
GNFDYWGQGTLVTVS S AS TKGPS VFPLAPS SKS TSGGTAAL
GCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSS
VVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCD
DR5(5E11) EIVLTQSPGTLSLSPGERATLSCRA SQS VS S SYLAWYQQKPG 139 pETR11480 YYCQQGTTHPITFGQGTKVEIKS S AS TKGPS VFPLAPS SKS TS
GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS
GLYSLSS VVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKS
CD
DRS (18F11)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 140 (28H1) GKGLEWVSAISGSGGSTYYADSVKGRPTISRDNSKNTLYLQ
VHCL MNS LRAEDAAVYYCAKGVRKKFDYWGQGTLVTVS S AS TK
2+2 GPSVFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNSGAL
TSGVHTFPAVLQS SGLYS LS SVVTVPS S SLGTQTYICNVNHK
PSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKA
LPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKSGGGG
SGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAAS
GI-TES SHAMS WVRQAPGKGLEWVS AIWASGEQYYADS VKG
RPTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVS S AS VAAPS VFIFPPSDEQLKSGTAS VVC LLNN
FYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTL
TLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
-208-N a me Amin() acid sequence SlQ II) N() DR5 (18F11) LC EIVLTQ S PGTLS LS PGER ATLS CR ASQS VS S S YLAWYQQKPG 141 QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFAV
YYCQQGQLPPITFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSG
TASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS
KDS TY S LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC
DR5(5E11)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 142 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRF1 ISRDNSKNTLYLQ
Fc knob MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
VHCL PS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
2+1 SGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYICNVNHKP
pETR10427 SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV
)¨( DKSRWQQGNVFS CS VMHEALHNHYTQKS LS LS PGGGGGS G
GGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGF
TFS S HAMS WVRQAPGKGLEWVS AIVVAS GEQYYADS VKGRF
TISRDNSKNTLYLQMNSLRAEDTAVYYCAKGWLGNFDYW
GQGTLVTVS SAS VAAPS VFIFPPSDEQLKSGTASVVCLLNNF
YPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS STLT
LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
DR5(5E11) EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 143 Fc hole GKGLEWVS AIS GS GGS TYYADS VKGRF1 ISRDNSKNTLYLQ
pETR10336 MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
PS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
SGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVS LS CAVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTV
DKSRWQQGNVFS CS VMHEALHNHYTQKS LS LS PGK
DR5(5E11)-FAP as above 143 (28H1) Fc knob VHCL
3+1 pETR10427
-209-N a me Amin() acid sequence SlQ II) N() DR5 (5E11)- EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 144 DR5(5E11) GKGLEWVSAISGSGGSTYYADSVKGRFIISRDNSKNTLYLQ
Fc hole MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
pETR10429 PS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNSGALT
3+11k. #4, SGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
VV.?

PAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTV
DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSG
GGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGF
TFS S YAMS WVRQAPGKGLEWVS AISGSGGS TYYADS VKGR
FI'ISRDNSKNTLYLQMNSLRAEDTAVYYCAKGVRVSFDYW
GQGTLVTVS S AS TKGPS VFPLAPS S KS TSGGTAALGCLVKDY
FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS
SLGTQTYICNVNHKPSNTKVDKKVEPKSCD
DR5(5E11)_Fc EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 145 knob GKGLEWVSAISGSGGSTYYADSVKGRFIISRDNSKNTLYLQ
Fab-Fab MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
Head-to-tail PS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNSGALT
2+1 SGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKP
pETR10662 SNTKVDKKVEPKSCGGGGSGGGGSEVQLLESGGGLVQPGG

TYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
KGVRVSFDYWGQGTLVTVS S AS TKGPSVFPLAPS SKS TSGG
TAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL
s=
YSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCD
KTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
DVSHEDPEVKFNVVYVDGVEVHNAKTKPREEQYNSTYRVVS
VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE
PQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQ
PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCS V
MHEALHNHYTQKSLSLSPGK
FAP (28H1) _Fc EVQLLESGGGLVQPGGSLRLSCAASGFTFS SHAMS WVRQAP 146 hole GKGLEWVSAIVVASGEQYYADSVKGRFTISRDNSKNTLYLQ
VHCL MNS LRAEDTAVYYCAKGWLGNFDYWGQGTLVTVS S AS VA
pETR10130 APS VFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDN
ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYA
CEVTHQGLSSPVTKSFNRGECDKTHTCPPCPAPELLGGPSVF
LFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNVVYVDGV
EVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK
VSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSL
SCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLV
SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
-210-N a me Amin() acid sequence SE() 11) N
DR5 (18F11)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 147 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
Fc knob MNS LRAEDAAVYYCAKGVRKKFDYWGQGTLVTVS S AS TK
VHCL GPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GAL
2+1 TSGVHTFPAVLQS SGLYS LS S VVTVPS S SLGTQTYICNVNHK
pETR9807 PS NTKVDKKVEPKS CDKTHTCPPCPAPELLGGPS VFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKA
LPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLV
KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT
VDKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGKS GGG
b, GSGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGS LRLS CA
ASGFTFS S HAMS WVRQAPGKGLEWVS AIWAS GEQYYADS V
KGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGWLGNF
DYWGQGTLVTVS SAS VAAPS VFIFPPSDEQLKSGTAS VVCLL
NNFYPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS S
TLTLSKADYEKHKVYACEVTHQGLS SPVTKSFNRGEC
DR5(18F11) Fc EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 148 hole GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
pETR9808 MNS LRAEDAAVYYCAKGVRKKFDYWGQGTLVTVS S AS TK
GPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GAL
TSGVHTFPAVLQS SGLYS LS S VVTVPS S SLGTQTYICNVNHK
PS NTKVDKKVEPKS CDKTHTCPPCPAPELLGGPS VFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKA
LPAPIEKTIS KAKGQPREPQVCTLPPSRDELTKNQVS LS CAVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTV
DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGK
DR5(18F11)-FAP EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 149 (28H1) GKGLEWVS AIS GS GGS TYYADS VKGRPTISRDNSKNTLYLQ
VHCL MNS LRAEDAAVYYCAKGVRKKFDYWGQGTLVTVS S AS TK
Fc knob GPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GAL
3+1 TSGVHTFPAVLQS SGLYS LS S VVTVPS S SLGTQTYICNVNHK
pETR10333 PS NTKVDKKVEPKS CDKTHTCPPCPAPELLGGPS VFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKA
F
LPAPIEKTIS KAKGQPREPQVYTLPPCRDELTKNQVSLWCLV
#,r VDKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGGGGGS
¨4 GGGGSGGGGSGGGGSEVQLLES GGGLVQPGGS LRLS CA AS
GPTFS S HAMS WVRQAPGKGLEWVS AIWAS GEQYYADS VKG
RPTISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWLGNFDY
WGQGTLVTVS SAS VAAPSVFIFPPSDEQLKSGTAS VVCLLNN
FYPREAKVQWKVDNALQSGNSQES VTEQDS KDS TYS LS STL
TLSKADYEKHKVYACEVTHQGLS SPVTKS FNRGEC

N a me Amin() acid sequence SlQ II) N() DR5 (18F11)- EVQLLES GGGLVQPGGS LRLS CA AS GFTFS S YAMS WVRQAP 150 DR5(18F11) GKGLEWVS AIS GS GGS TYYADS VKGRF1 IS RDNS KNTLYLQ
Fc hole MNS LRAEDAAVYYCAKGVRKKFDYWGQGTLVTVS S AS TK
pETR10288 GPS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GAL
TSGVHTFPAVLQS SGLYS LS S VVTVPS S SLGTQTYICNVNHK
PS NTKVDKKVEPKS CDKTHTCPPCPAPELLGGPS VFLFPPKP
KDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKA
LPAPIEKTIS KAKGQPREPQVCTLPPSRDELTKNQVS LS CAVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTV
DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGGGGGS G
GGGSGGGGS GGGGSEVQLLESGGGLVQPGGS LRLS CA AS GF
TFS S YAMS WVRQAPGKGLEWVS AIS GS GGS TYYADS VKGR
Fl IS RDNS KNTLYLQMNS LRAEDAAVYYCAKGVRKKFDYW
GQGTLVTVS S AS TKGPS VFPLAPS S KS TS GGTAALGCLVKDY
FPEPVTVSWNSGALTSGVHTFPAVLQS S GLYS LS S VVTVPS S
SLGTQTYICNVNHKPSNTKVDKKVEPKSCD
FAP (28H1) _Fc EVQLLES GGGLVQPGGS LRLS CA AS GFTFS S HAMS WVRQAP 159 hole GKGLEWVS AIVVASGEQYYADS VKGRFTISRDNS KNTLYLQ
VHCL MNS LRAEDTAVYYCAKGWLGNFDYWGQGTLVTVS SAS VA
pETR10130 APS VFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDN
ALQSGNSQES VTEQDS KDS TYS LS STLTLSKADYEKHKVYA
CEVTHQGLS SPVTKS FNRGECDKTHTCPPCPAPELLGGPS VF
LFPPKPKDTLMISRTPEVTCVVVDVS HEDPEVKFNVVYVDGV
EVHNAKTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCK

SCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLV
SKLTVDKS RWQQGNVFS C SVMHEALHNHYTQKS LS LS PGK
FAP (28H1) EIVLTQS PGTLS LS PGERATLS CRASQ SVS RS YLAWYQQKPG 160 pETR9537 YYCQQGQVIPPTFGQGTKVEIKS SAS TKGPS VFPLAPS S KS TS
GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS S
GLYS LS S VVTVPS S SLGTQTYICNVNHKPSNTKVDKKVEPKS
CD
DR5 (5 Ell )-Fc EVQLLESGGGLVQPGGSLRLSCAASGFTFS S YAMS WVRQAP 161 knob GKGLEWVS AIS GS GGS TYYADS VKGRF1 IS RDNS KNTLYLQ
MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
PS VFPLAPS S KS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
SGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVS LWCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV
DKS RWQQGNVFS CS VMHEALHNHYTQKS LS LS PGK
DRS (5 Ell )_LC EIVLTQS PGTLS LS PGERATLS CRASQ SVS S SYLAWYQQKPG 162 pETR9044 QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTIS RLEPEDFA V
YYCQQGTTHPITFGQGTKVEIKRTVAAPS VFIFPPSDEQLKSG
TASVVCLLNNFYPREAKVQWKVDNALQS GNSQES VTEQDS
KDS TY S LS STLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGEC

Name Amino acid ,,cgticiice SEQ 11) N() DR5 (5E11) EVQLLES GGGLVQPGGS LRLS CA AS GFTFS S YAMS WVRQAP 262 FAP(4B9) GKGLEWVS AIS GS GGS TYYADS VKGRF1 IS RDNS KNTLYLQ
VHCL MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
pETR11060 PS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNS GALT
SGVHTFPAVLQS SGLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
i=r DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNS TYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV
DKS RWQQ GNVFS CS VMHEALHNHYTQK S LS L S PGGGGGS G
GGGSGGGGS GGGGS EV QLLES GGGLVQPGGS LRLS CA A S GF
TFS S YAMS WVRQAPGKGLEWVS AIIGS GA S TYYADS VKGRF
TISRDNSKNTLYLQMNS LRAEDTAVYYCAKGWFGGFNYWG
QGTLVTVS S AS VA APS VFIFPPSDEQLKSGTAS VVCLLNNFY
PREAKVQWKVDNALQ SGNS QES VTEQDSKDS TYS LS S TLTL
SKADYEKHKVYACEVTHQGLS SPVTKSFNRGEC
FAP(4B9) EIVLTQ S PGTLS LS PGER ATLS CRA SQS VT S S YLAWYQQKPG 263 pETR10020 VYYCQQGIMLPPTFGQGTKVEIKS S AS TKGPS VFPLAPS SKS T
SGGTAALGCLVKDYFPEPVTVS WNSGALTS GVHTFPAVLQS
SGLYS LS S VVTVPS S SLGTQTYICNVNHKPSNTKVDKKVEPK
SCD

5E11 (VLCL)-Fc- QAPRLLIYGAS SRATGIPDRFS GS GS GTDFTLTISRLEPEDFA V
28H1 (VHCH1) YYCQQGTTHPITFGQGTKVEIKRTVAAPS VFIFPPSDEQLKSG
TA S VVC LLNNFYPREAKVQWKVDNALQ S GNS QES VTEQDS
KDS TYS LS S TLTLSKADYEKHKVYACEVTHQGLS SPVTKSF
NRGECDKTHTCPPCPAPELLGGPS VFLFPPKPKDTLMISRTPE
VTCVVVDVS HEDPEVKFNWYVDGVEVHNAKTKPREEQYN
STYRVVS VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS K
AKGQPREPQVYTLPPSRDELTKNQVS LTCLVKGFYPSDIAVE
WES NGQPENNYKTTPPVLDS DGS FFLYSKLTVDKSRWQQG
NVFSCS VMHEALHNHY TQKS LS LS PGGGGGS GGGGSGGGG
SGGGGS EVQ LLES GGGLVQPGGS LRLS C AA S GFTFS SHAMS
WVRQAPGKGLEWVS AIWA S GEQYYADS VKGRFTISRDNSK
NTLYLQMNS LRAEDTAVYYCAKGWLGNFDYWGQGTLVTV
S S A S TKGPS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQS SGLYS LS S VVTVPS S S LGTQTYI

MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
PS VFPLAPS SKS TSGGTAALGCLVKDYFPEPVTVS WNS GALT
SGVHTFPAVLQS SGLY S LS S VVTVPS S SLGTQTYICNVNHKP

Name Amino acid wquence SEQ
11) N() PS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
SGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKSRWQQGNVFS CS VMHEALHNHYTQKS LS LSPGGGGGS G
GGGSGGGGS GGGGSEIVLTQSPGTLS LSPGERATLS CRAS QS
VSRS YLAWYQQKPGQAPRLLIIGAS TRATGIPDRFS GS GS GT

APS VFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDN
ALQSGNSQES VTEQDSKDS TYS LS STLTLSKADYEKHKVYA

MNS LRAEDTAVYYCAKGWLGNFDYWGQGTLVTVS S AS TK
GPS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS WNS GAL
TSGVHTFPAVLQS SGLYS LS S VVTVPS S SLGTQTYICNVNHK

(VLCH1) MNS LRAEDTAVYYCAKGVRVSFDYWGQGTLVTVS S AS TKG
PS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS WNS GALT
SGVHTFPAVLQS S GLY S LS S VVTVPS S SLGTQTYICNVNHKP
SNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
TKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKAL
GAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK
GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFELYSKLTV
DKSRWQQGNVFS CS VMHEALHNHYTQKS LS LSPGGGGGS G
GGGSGGGGS GGGGSEIVLTQSPGTLS LSPGERATLS CRAS QS
VSRS YLAWYQQKPGQAPRLLIIGAS TRATGIPDRFS GS GS GT

KGPS VFPLAPS SKS TS GGTAALGCLVKDYFPEPVTVS WNS G
ALTSGVHTFPAVLQS S GLYS LS S VVTVPS S S LGTQTYICNVN

28H1 (VHCL) GKGLEWVSAIWASGEQYYADSVKGREI ISRDNSKNTLYLQM
NS LRAEDTAVYYCAKGWLGNFDYWGQGTLVTVS SAS VAAP
S VFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDNALQ
SGNSQES VTEQDSKDS TYS LS STLTLSKADYEKHKVYACEVT

6. Amino acid sequences of Fc domain and constant light chains Name Amin() ticid sequence SEQ II) NO
hu Fc _wt ASTKGPS VEPLAPS SKS ISGGIAALGCLVKD YEPEPVIV S WN SGAL 151 TSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTK
VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLEPPKPKDTLMISRTP
EVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR
VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP
QVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY
KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHY
TQKSLSLSPGK
hu ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGAL 152 Fc_P329G/LALA TSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTK
VDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLEPPKPKDTLMISRT
PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTY
RVVSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTISKAKGQPRE
PQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY
KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHY
TQKSLSLSPK
hu kappa light RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNEYPREAKVQWKVDN 153 chain ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVT
HQGLSSPVTKSFNRGEC
hu lambda light GQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADS 154 chain SPVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTH
EGSTVEKTVAPTECS
7. Amino acid sequences of DR5 binders derived from rabbit immunization Clone DR5TAA-0005 (Alias: clone 039) Heavy chain VH
qsleesggrlvtpgtpldtctasgfslssaymswvrqapgkglewigyiysgsgstwyaswvkgrftisktsttvdlki tspttedtatyfc argystmgdlwgpgtivtvss (SEQ ID NO.: 41) gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntkv dktvapstcskptcpppellggpsvfifppkpkddmisrtpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvstl piahqdwligkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekng kaednykt tpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = sayms (SEQ ID NO.: 43) CDR2 = yiysgsgstwyaswvkg (SEQ ID NO.: 44) CDR3 = gystmgdl (SEQ ID NO.: 45) Light Chain VL
qvltqtpspvsaavggtvtincqasqsvynnrlawyqqkpgqppklliylastlasgvpsrfkgsgsgtqftltisdlq cddaatyycagg ysgninafgggtevvvk (SEQ ID NO.: 46) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 47) CDR1 = qasqsvynnrla (SEQ ID NO.: 48) CDR2 = lastlas (SEQ ID NO.: 49) CDR3 = aggysgnina (SEQ ID NO.: 50) Clone DR5TAA-0006 (Alias: clone 058) Heavy chain VH
qsleesggrlvtpgtpldtctasgfslssnhmswvrqapgkglewigyiyagsgsayyaswakgrftisrtsttvd1km tslttedtatyfc agdagssywefnlwgpgtivtvss (SEQ ID NO.: 51) gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntkv dktvapstcskptcpppellggpsvfifppkpkddmisrtpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvstl piahqdwligkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekng kaednykt tpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = snhms (SEQ ID NO.: 52) CDR2 = yiyagsgsayyaswakg (SEQ ID NO.: 53) CDR3 = dagssywefnl (SEQ ID NO.: 54) Light chain VL
lvmtqtpsstsepvggtvtikcqasqsigsslswyqqkpgqppklliyhastlasgvpsrfsgsrsgiqttltisgvqc ddaatyyclgvad arrddgfafgggtevvvk (SEQ ID NO.: 55) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 56) CDR1 = qasqsigssls (SEQ ID NO.: 57) CDR2 = hastlas (SEQ ID NO.: 58) CDR3 = lgvadarrddgfa (SEQ ID NO.: 59) Clone DR5TAA-0010 (Alias: clone 481) Heavy chain VH
qsleesggrlvkpdetltltctvsgfslssnaiswvrqapgmglewigiigssgytyyaswakgrftvsktsttvdlei aspttedtatyfcar gysgasdysfnlwgpgtivtvss (SEQ ID NO.: 60) gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntkv dktvapstcskptcpppellggpsvfifppkpkddmisrtpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvstl piahqdwligkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekng kaednykt tpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = snais (SEQ ID NO.: 61) CDR2 = iigssgytyyaswakg (SEQ ID NO.: 62) CDR3 = gysgasdysfnl (SEQ ID NO.: 63) Light chain VL
aydmtqtpdsvevavggtvtikcqasqtigdalawyqqkpgqrpnlliyrtstlasgvpsrfsgsgsgthfdtisgvec adaatyycqqg atynnvintfgggtevvvk (SEQ ID NO.: 64) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 47) CDR1 = qasqtigdala (SEQ ID NO.: 65) CDR2 = rtstlas (SEQ ID NO.: 66) CDR3 = qqgatynnylnt (SEQ ID NO.: 67) Clone DR5TAA-0013 (Alias: clone 298) Heavy chain VH
Qsleesggrlvtpgtpldtctasgftissyhmswvrqapgkglewigyiyagsastwyaswvkgrftisktsttvd1km tslttedtatyf cardagssywefnlwgpgtivtvss (SEQ ID NO.: 68) gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntkv dktvapstcskptcpppellggpsvfifppkpkddmisrtpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvstl piahqdwligkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekng kaednykt tpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = syhms (SEQ ID NO.: 69) CDR2 = yiyagsastwyaswvkg (SEQ ID NO.: 70) CDR3 = dagssywefnl (SEQ ID NO.: 54) Light chain VL
Lvmtqtpsstsepvggtvtikcqasqsigsslswyqqtpgqppklliytasslassvpkrfsgsrsgtqftltisgvqc adaatyyclgidd vrrddgfafgggtevvvk (SEQ ID NO.: 71) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 47) CDR1 = qasqsigssls (SEQ ID NO.: 57) CDR2 = tasslas (SEQ ID NO.: 72) CDR3 = lgiddvrrddgfa (SEQ ID NO.: 73) Clone DR5TAA-0019 (Alias: clone 461) Heavy chain VH
Qsveesggrlvtpgtpldtctvsgfslsnyamswvrqapgkglewigiisssgttyyaswakgrftisktsttvd1kvt spttedtatyfca retyygysyaaglwgpgtivtvss (SEQ ID NO.: 74) gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntkv dktvapstcskptcpppellggpsvfifppkpkddmisrtpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvstl piahqdwligkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekng kaednykt tpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = nyams (SEQ ID NO.: 75) CDR2 = iisssgttyyaswakg (SEQ ID NO.: 76) CDR3 = etyygysyaagl (SEQ ID NO.: 77) Light chain VL
Alvmtqtpssvsaavggtvtincqasqniysnlawfqqkpgqppklliyetsklasgvpsrfsgsgsgteftltisdle cddaatyycqss whsistdcafgggtevvvk (SEQ ID NO.: 78) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 47) CDR1 = qasqniysnla (SEQ ID NO.: 79) CDR2 = etsklas (SEQ ID NO.: 80) CDR3 = qsswhsistdca (SEQ ID NO.: 81) Clone DR5TAA-0016 (Alias: clone 422) Heavy chain VH
Qsleesggrlvkpdetldtctvsgfslnnyamswvrqapgkglewigminkygtkyyatwtkgratisktstddleits pttedtatyfc arvryagddyaewldvwgqgilvtvss (SEQ ID NO.: 82) gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntkv dktvapstcskptcpppellggpsvfifppkpkddmisrtpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvstl piahqdwligkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekng kaednykt tpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = nyams (SEQ ID NO.: 75) CDR2 = minkygtkyyatwtkg (SEQ ID NO.: 83) CDR3 = vryagddyaewldv (SEQ ID NO.: 84) Light chain VL
Adivmtqtaspvsaavggtvtincqasqsisssyvswyqqkpgqppklliykastlasgvpsrfsgsgsgtqlsltirg vqcddaatyyc lygysdvssseyvfgggtevvvr (SEQ ID NO.: 85) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 47) CDR1 = qasqsisssyvs (SEQ ID NO.: 86) CDR2 = kastlas (SEQ ID NO.: 28) CDR3 = lygysdvssseyv (SEQ ID NO.: 87) Clone DR5TAA-0011 (Alias: clone 174) Heavy chain VH
Qsveesggrlvtpgtpldtctvsgfsisryamiwvrqapgegleyigfitsdssayyaswakgrftisktsttvd1kmt spttedtatyfca rytysdgtdlwgpgtivtvss (SEQ ID NO.: 88) Gqpkapsvfplapccgdtpsstvtlgclvkgylpepvtvtwnsgtltngvrtfpsvrqssglyslssvvsvtsssqpvt cnvahpatntk vdktvapstcskptcpppellggpsvfifppkpkddmisdpevtcvvvdvsqddpevqftwyinneqvrtarpplreqq fnstirvvs dpiahqdwlrgkefkckvhnkalpapiektiskargqplepkvytmgppreelssrsvsltcmingfypsdisvewekn gkaedny kttpavldsdgsyflynklsvptsewqrgdyftcsvmhealhnhytqksisrspgk (SEQ ID NO.: 42) CDR1 = ryami (SEQ ID NO.:17) CDR2 = fitsdssayyaswakg (SEQ ID NO.: 25) CDR3 = ytysdgtdl (SEQ ID NO.: 19) Light Chain VL
Adivmtqtpasvsepvggtvtikcqasqsistylswyqqkpgqppkrliykastlasgvpsrfkgsgsgtdftltirdl ecadaatyycq pnsgiatygaafgggtevvvk (SEQ ID NO.: 89) Ckappa gdpvaptvlifppaadqvatgtvtivcvankyfpdvtvtwevdgttqttgiensktpqnsadctynlsstltltstqyn shkeytckvtqgt tsvvqsfnrgdc (SEQ ID NO.: 47) CDR1 = qasqsistyls (SEQ ID NO.: 27) CDR2 = kastlas (SEQ ID NO. :28) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) 8. Amino acid sequences of chimerized variant of rabbit Mab DR5TAA-0011 Clone DR5TAA-0052 (chimeric variant) Heavy chain VH
Qsveesggrlvtpgtpldtetvsgfsisryamiwvrqapgegleyigfitsdssayyaswakgrftisktsttvd1kmt spttedtatyfea rytysdgtdlwgpgtivtvss (SEQ ID NO.: 90) Astkgpsvfplapsskstsggtaalgelvkdyfpepvtvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtq tyienvnhkps ntkvdkkvepksedkthteppepapeaaggpsvflfppkpkddmisdpevtevvvdvshedpevkfnwyvdgvevhnak tkpr eeqynstyrvvsyltvlhqdwingkeykekvsnkalgapiektiskakgqprepqvytlppsrdeltknqvsltelvkg fypsdiave wesngqpennykttppvldsdgsfflyskltvdksrwqqgnvfsesvmhealhnhytqks1s1spgk (SEQ ID
NO.: 91) CDR1 = ryami (SEQ ID NO.: 17) CDR2 = fitsdssayyaswakg (SEQ ID NO.: 25) CDR3 = ytysdgtdl (SEQ ID NO.: 19) Light chain VH
Adivmtqtpasvsepvggtvtikeqasqsistylswyqqkpgqppkrliykastlasgvpsrfkgsgsgtdftltirdl esadaatyycq pnsgiatygaafgggtevvvk (SEQ ID NO.: 92) Ckappa Rtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstldskadye khkvyacev thqglsspvtksfnrgec (SEQ ID NO.: 93) CDR1 = qasqsistyls (SEQ ID NO.: 27) CDR2 = kastlas (SEQ ID NO.: 28) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) 9. Sequences of humanized variants of rabbit Mab DR5TAA-0011 Variant HC LC
variant variant DR5TAA-0066 humanized VH7 VL3 DR5TAA-0067 humanized VH7 VL15 DR5TAA-0068 Humanized VH17 VL10 DR5TAA-0071 Humanized VH17 VL15 DR5TAA-0072 Humanized VH17 VL2 DR5TAA-0073 Humanized VH17 VL3 DR5TAA-0074 Humanized VH7 VL10 DR5TAA-0075 Humanized VH7 VL11 Humanized variants Heavy Chain VH
Evqlvetgggliqpggslrlscaasgftvsryamiwvrqapgkgleyigfitsdgstyyadsakgrftisrdnskntly lqmnslraedta vyyearytysdgtd1wgrgtivtvss (SEQ ID NO.: 23) astkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtq tyicnvnhkpsn tkvdkkvepkscdkthtcppcpapeaaggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkfnwyvdgvevhna ktkpre eqynstyrvvsyltvlhqdwingkeykckvsnkalgapiektiskakgqprepqvytlppsrdeltknqvsltclvkgf ypsdiavew esngqpennykttppvldsdgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgk constant chain (SEQ
ID NO.: 91) CDR1 = ryami (SEQ ID NO.: 17) CDR2 = fitsdgstyyadsakg (SEQ ID NO.: 18) CDR3 = ytysdgtdl (SEQ ID NO.: 19) VH
qvqlvesggglvqpggslrlscsasgfsisryamiwvrqapgkgleyvgfitsdssayyaswakgrftisrdnskntly lqmnslraedt avyycarytysdgtd1wgqgtivtvss (SEQ ID NO.: 26) astkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtq tyicnvnhkpsn tkvdkkvepkscdkthtcppcpapeaaggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkfnwyvdgvevhna ktkpre eqynstyrvvsyltvlhqdwingkeykckvsnkalgapiektiskakgqprepqvytlppsrdeltknqvsltclvkgf ypsdiavew esngqpennykttppvldsdgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgk (SEQ ID
NO.: 91) CDR1 = ryami (SEQ ID NO.: 17) CDR2 = fitsdssayyaswakg (SEQ ID NO.: 25) CDR3 = ytysdgtdl (SEQ ID NO.: 19) Humanized variants Light Chain VL
Diqmtqspstlsasvgdrvtitcrasqsistylswyqqkpgkapkrliykasslasgvpsrfsgsgsgteftltisslq pddaatyycqpns giatygaafgggtkveik (SEQ ID NO.: 32) Ckappa Rtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstldskadye khkvyacev thqglsspvtksfnrgec (SEQ ID NO.: 93) CDR1 = rasqsistyls (SEQ ID NO.: 20) CDR2 = kasslas (SEQ ID NO. :21) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) VL
Diqmtqspsslsasvgdrvtitcrasqsistylswyqqkpgkapkrliykastlasgvpsrfsgsgsgtdfdtisslqp edaatyycqpns giatygaafgggtkveik (SEQ ID NO.: 30) Ckappa Rtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstldskadye khkvyacev thqglsspvtksfnrgec (SEQ ID NO.: 93) CDR1 = rasqsistyls (SEQ ID NO. :20) CDR2 = kastlas (SEQ ID NO.: 28) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) VL
diqmtqspsslsasvgdrvtitcrasqsistylswyqqkpgqppkrliykastlasgvpsrfsgsgsgtdfdtisslqp edfatyycqpnsg iatygaafgggtkveik (SEQ ID NO.: 31) Ckappa rtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstldskadye khkvyacevt hqglsspvtksfnrgec (SEQ ID NO.: 93) CDR1 = rasqsistyls (SEQ ID NO. :20) CDR2 = kastlas (SEQ ID NO.: 28) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) VL
diqmtqspsslsasvgdrvtiteqasqsistylswyqqkpgqppkrliykastlasgvpsrfsgsgsgtdfdtisslqp edfatyycqpns giatygaafgggtkveik (SEQ ID NO.: 29) Ckappa rtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstldskadye khkvyacevt hqglsspvtksfnrgec (SEQ ID NO.: 93) CDR1 = qasqsistyls (SEQ ID NO.: 27) CDR2 = kastlas (SEQ ID NO.: 28) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) VL
Adiqmtqspstlsasvgdrvtiterasqsistylswyqqkpgkapkrliykasslasgvpsrfsgsgsgteftltissl qpddaatyycqpn sgiatygaafgggtkveik (SEQ ID NO.: 24) Ckappa Rtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstldskadye khkvyacev thqglsspvtksfnrgec (SEQ ID NO.: 93) CDR1 = rasqsistyls (SEQ ID NO.: 20) CDR2 = kasslas (SEQ ID NO.: 21) CDR3 = qpnsgiatygaa (SEQ ID NO.: 22) 10. Amino acid sequences of chimeric bispecific constructs containing rabbit-derived DR5-binder Construct DR5TAA-0061 (1+1 chimeric Crossmab containing 4B9 and DR5TAA-0011) LC (DR5) adivmtqtpasvsepvggtvtikcgasgsistylswyqqkpgqppkrliykastlasgvpsrfkgsgsgtdftltir dlecadaatyycqpnsgiatygaafgggtevvvkrtvaapsvfifppsdeqlksgtasvvcllnnfypreakvqwkv dnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacev thqglsspvtksfnrgec (SEXPE)Na:280) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvtssylawyqqkpgqaprllinvgsrratgipdrfsgsgsgtdftltis rlepedfavyycqggimlpptfgqgtkveikssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsg altsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvnhkps ntkvdkkvepkscd (SEXPE)Na:281) HC (DR5) gsveesggrlvtpgtpltltetvsgfsisryamiwvrqapgegleyigfitsdssayyaswakgrftisktsttvd1 kmtspttedtatyfearytysdgtd1wgpgtivtvssastkgpsvfplapsskstsggtaalgelvkdyfpepvtvs wnsgaltsgvhtfpavlgssglyslssvvtvpssslgtqtyienvn hkpsntkvdkkvepksedkthteppepapeaaggpsvflfppkpkdtlmisrtpevtevvvdvshedpevkfnwyvd gvevhnaktkpreegynstyrvvsvltvlhqdwingkeykekvsnkalgapiektiskakgqprepqvctlppsrde ltknqvs1scavkgfypsdiavewesnggpennykttppvldsdgs fflvskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgk (SWIDNa:282) Crossed HC (FAP) evq1lesggglvqpggslrlscaasgftfssyamswvrqapgkglewvsaiigsgastyyadsvkgrftisrdnskn tlylqmnslraedtavyyeakgwfggfnywgqgtivtvssasvaapsvfifppsdeqlksgtasvvellnnfyprea kvqwkvdnalgsgnsgesvtegdskdstyslsstltlskadyekhk vyacevthqglsspvtksfnrgeedkthteppepapeaaggpsvflfppkpkdtlmisrtpevtevvvdvshedpev kfnwyvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykekvsnkalgapiektiskakgqprepqvyt lpperdeltknqvslwelvkgfypsdiavewesnggpennykttpp vldsdgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgk (SWIDNXI:283) Construct DR5TAA-0032 (2+2 chimeric CrossMab containing 28H1 and DR5TAA-0005) LC (DR5) aqvltqtpspvsaavggtvtinegasgsvynnrlawyqqkpgqppklliylastlasgvpsrfkgsgsgtqftltis dlqeddaatyycaggysgninafgggtevvvkrtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdn algsgnsgesvtegdskdstyslsstltlskadyekhkvyacevth gglsspvtksfnrgee (SEQIDNa:284) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgelvkdyfpepvtvswnsg altsgvhtfpavlgssglyslssvvtvpssslgtqtyienvnhkps ntkvdkkvepkscd (SWIDNa:285) HC (DR5 - crossed FAP) qsleesggrlvtpgtpltlictasgfslssaymswvrqapgkglewigyiysgsgstwyaswvkgrftisktsttvdlk itspttedtatyfcargystmg dlwgpgtivtvssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsgaltsgvhtfpavlqssglysls svvtvpssslgtqtyienvnh kpsntkvdkkvepkscdkthtcppcpapellggpsvflfppkpkddmisrtpevtcvvvdvshedpevkfnwyvdgvev hnaktkpreeqyn styrvvsyltvlhqdwingkeykekvsnkalpapiektiskakgqprepqvytlppsrdeltknqvsltclvkgfypsd iavewesngqpennyktt ppvldsdgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgksggggsggggsggggsggggsevq1l esggglvqpggslrls caasgftfsshamswvrqapgkglewvsaiwasgeqyyadsvkgrftisrdnskntlylqmnslraedtavyycakgwl gnfdywgqgtivtvs sasvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkvdnalqsgnsqesvteqdskdstyslsstlilskad yekhkvyacevthqglssp vtksfnrgec (SEQ ID NO.: 286) Construct DR5TAA-0033 (2+2 chimeric CrossMab containing 28H1 and DR5TAA-0011) LC (DR5) adivmtqtpasvsepvggtvtikegasgsistylswyqqkpgqppkrliykastlasgvpsrfkgsgsgtdftltir dlecadaatyycqpnsgiatygaafgggtevvvkrtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkv dnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacev thqglsspvtksfnrgec (SWIDNXI:287) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsg altsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvnhkps ntkvdkkvepkscd (SEXPE)NXI:288) HC (DR5 - crossed FAP) gsveesggrlvtpgtpltltctvsgfsisryamiwvrqapgegleyigfitsdssayyaswakgrftisktsttvd1 kmtspttedtatyfcarytysdgtd1wgpgtivtvssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvs wnsgaltsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvn hkpsntkvdkkvepkscdkthtcppcpapellggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkfnwyvd gvevhnaktkpreegynstyrvvsvltvlhqdwingkeykckvsnkalpapiektiskakgqprepqvytlppsrde ltknqvsltclvkgfypsdiavewesnggpennykttppvldsdgs fflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgksggggsggggsggggsggggsevq1lesggglv qpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmnslraed tavyycakgwlgnfdywgqgtivtvssasvaapsvfifppsdeqlk sgtasvvc11nnfypreakvqwkvdnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevthqglss pvtksfnrgec (SEXPE)NNI:289) Construct DR5TAA-0034 (2+2 chimeric CrossMab containing 28H1 and DR5TAA-0013) LC (DR5) alvmtqtpsstsepvggtvtikcgasgsigsslswyqqtpgqppklliytasslassvpkrfsgsrsgtqftltisg vqcadaatyyclgiddvrrddgfafgggtevvvkrtvaapsvfifppsdeqlksgtasvvcllnnfypreakvqwkv dnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacev thqglsspvtksfnrgec (SEXPE)NNI:290) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsg altsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvnhkps ntkvdkkvepkscd (SEXPE)NXI:291) HC (DR5 - crossed FAP) gsleesggrlvtpgtpltltctasgftissyhmswvrqapgkglewigyiyagsastwyaswvkgrftisktsttvd lkmtslttedtatyfcardagssywefnlwgpgtivtvssastkgpsvfplapsskstsggtaalgclvkdyfpepv tvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtqtyic nvnhkpsntkvdkkvepkscdkthtcppcpapellggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkfnw yvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykckvsnkalpapiektiskakgqprepqvytlpps rdeltknqvsltclvkgfypsdiavewesnggpennykttppvlds dgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgksggggsggggsggggsggggsevq1lesgg glvqpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmnslr aedtavyycakgwlgnfdywgqgtivtvssasvaapsvfifppsde qlksgtasvvcllnnfypreakvqwkvdnalqsgnsgesvteqdskdstyslsstltlskadyekhkvyacevthqg lsspvtksfnrgec (SEXPE)NXI:292) Construct DR5TAA-0035 (2+2 chimeric CrossMab containing 28H1 and DR5TAA-0016) LC (DRS) adivmtqtaspvsaavggtvtincgasgsisssyvswyqqkpgqppklliykastlasgvpsrfsgsgsgtqlslti rgvqcddaatyyclygysdvssseyvfgggtevvvrrtvaapsvfifppsdeqlksgtasvvcllnnfypreakvqw kvdnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyac evthqglsspvtksfnrgec (SEXPDMI:293) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsg altsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvnhkps ntkvdkkvepkscd (SEXPDMI:294) HC (DR5 ¨ crossed FAP) gsleesggrlvkpdetltltctvsgfslnnyamswvrqapgkglewigminkygtkyyatwtkgratisktsttldl eitspttedtatyfcarvryagddyaewldvwgggilvtvssastkgpsvfplapsskstsggtaalgclvkdyfpe pvtvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtqty icnvnhkpsntkvdkkvepkscdkthtcppcpapellggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkf nwyvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykckvsnkalpapiektiskakgqprepqvytlp psrdeltknqvsltclvkgfypsdiavewesngqpennykttppvl dsdgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgksggggsggggsggggsggggsevq1les ggglvqpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmns lraedtavyycakgwlgnfdywgqgtivtvssasvaapsvfifpps deqlksgtasvvcllnnfypreakvqwkvdnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevth qglsspvtksfnrgec (SEQIDNXI:295) Construct DR5TAA-0036 (2+2 chimeric CrossMab containing 28H1 and DR5TAA-0019) LC (DR5) alvmtqtpssvsaavggtvtincgasqnlysnlawfqqkpgqppklliyetsklasgvpsrfsgsgsgteftltisd lecddaatyycgsswhsistdcafgggtevvvkrtvaapsvfifppsdeqlksgtasvvcllnnfypreakvqwkvd nalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevt hqglsspvtksfnrgec (SEQID NO.: 296) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsg altsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvnhkps ntkvdkkvepkscd (SEXPDPOD.: 297) HC (DR5 - crossed FAP) gsveesggrlvtpgtpltltctvsgfslsnyamswvrqapgkglewigiisssgttyyaswakgrftisktsttvd1 kvtspttedtatyfcaretyygysyaaglwgpgtivtvssastkgpsvfplapsskstsggtaalgclvkdyfpepv tvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtqtyic nvnhkpsntkvdkkvepkscdkthtcppcpapellggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkfnw yvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykckvsnkalpapiektiskakgqprepqvytlpps rdeltknqvsltclvkgfypsdiavewesngqpennykttppvlds dgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgksggggsggggsggggsggggsevq1lesgg glvqpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmnslr aedtavyycakgwlgnfdywgqgtivtvssasvaapsvfifppsde qlksgtasvvcllnnfypreakvqwkvdnalqsgnsgesvteqdskdstyslsstltlskadyekhkvyacevthqg lsspvtksfnrgec (SEXPDMI:298) 11. Amino acid sequences of bispecific constructs containing humanized rabbit-derived DR5-binder Construct DR5TAA-0117 (2+2 humanized CrossMab containing 28H1 and DR5TAA-0067) LC (DR5) adigmtgspstlsasvgdrvtiterasgsistylswyqqkpgkapkrliykasslasgvpsrfsgsgsgteftltis slqpddaatyycqpnsgiatygaafgggtkveikrtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkv dnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacev thqglsspvtksfnrgec (SEQ ID NO.: 299) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgelvkdyfpepvtvswnsg altsgvhtfpavlgssglyslssvvtvpssslgtqtyienvnhkps ntkvdkkvepkscd (SWIDNXI:300) HC (DR5 ¨ crossed FAP) evqlvetgggliqpggslrlscaasgftvsryamiwvrqapgkgleyigfitsdgstyyadsakgrftisrdnsknt lylqmnslraedtavyyearytysdgtd1wgrgtivtvssastkgpsvfplapsskstsggtaalgelvkdyfpepv tvswnsgaltsgvhtfpavlgssglyslssvvtvpssslgtqtyie nvnhkpsntkvdkkvepksedkthteppepapeaaggpsvflfppkpkdtlmisrtpevtevvvdvshedpevkfnw yvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykekvsnkalgapiektiskakgqprepqvytlpps rdeltknqvsltelvkgfypsdiavewesnggpennykttppvlds dgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgggggsggggsggggsggggsevq1lesgggl vqpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmnslrae dtavyyeakgwlgnfdywgqgtivtvssasvaapsvfifppsdeql ksgtasvvellnnfypreakvqwkvdnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevthqgls spvtksfnrgec (SEQ ID NO.: 301) Construct DR5TAA-0118 (2+2 humanized CrossMab containing 28H1 and DR5TAA-0071) LC (DR5) adigmtgspstlsasvgdrvtiterasgsistylswyqqkpgkapkrliykasslasgvpsrfsgsgsgteftltis slqpddaatyycqpnsgiatygaafgggtkveikrtvaapsvfifppsdeqlksgtasvvellnnfypreakvqwkv dnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacev thqglsspvtksfnrgec (SEQ ID NO.: 302) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgelvkdyfpepvtvswnsg altsgvhtfpavlgssglyslssvvtvpssslgtqtyienvnhkps ntkvdkkvepkscd (SWIDNXI:303) HC (DR5 - crossed FAP) qvglvesggglvqpggslrlscsasgfsisryamiwvrqapgkgleyvgfitsdssayyaswakgrftisrdnsknt lylqmnslraedtavyyearytysdgtd1wgqgtivtvssastkgpsvfplapsskstsggtaalgelvkdyfpepv tvswnsgaltsgvhtfpavlgssglyslssvvtvpssslgtqtyie nvnhkpsntkvdkkvepksedkthteppepapeaaggpsvflfppkpkdtlmisrtpevtevvvdvshedpevkfnw yvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykekvsnkalgapiektiskakgqprepqvytlpps rdeltknqvsltelvkgfypsdiavewesnggpennykttppvlds dgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgggggsggggsggggsggggsevq1lesgggl vqpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmnslrae dtavyycakgwlgnfdywgqgtivtvssasvaapsvfifppsdeql ksgtasvvc11nnfypreakvqwkvdnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevthqgls spvtksfnrgec (SWIE)1\10.:304) Construct DR5TAA-0119 (2+2 humanized CrossMab containing 28H1 and DR5TAA-0075) LC (DR5) digmtgspsslsasvgdrvtitcgasgsistylswyqqkpgqppkrliykastlasgvpsrfsgsgsgtdftltiss lqpedfatyycqpnsgiatygaafgggtkveikrtvaapsvfifppsdeqlksgtasvvcllnnfypreakvqwkvd nalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevt hqglsspvtksfnrgec (SW11)1\1(1:305) Crossed LC (FAP) eivltqspgt1s1spgeratlscrasgsvsrsylawyqqkpgqaprlliigastratgipdrfsgsgsgtdftltis rlepedfavyycqqgqvipptfgqgtkveikssastkgpsvfplapsskstsggtaalgclvkdyfpepvtvswnsg altsgvhtfpavlqssglyslssvvtvpssslgtqtyicnvnhkps ntkvdkkvepkscd (SW11)1\1(1:306) HC (DR5 - crossed FAP) evqlvetgggliqpggslrlscaasgftvsryamiwvrqapgkgleyigfitsdgstyyadsakgrftisrdnsknt lylqmnslraedtavyycarytysdgtd1wgrgtivtvssastkgpsvfplapsskstsggtaalgclvkdyfpepv tvswnsgaltsgvhtfpavlqssglyslssvvtvpssslgtqtyic nvnhkpsntkvdkkvepkscdkthtcppcpapeaaggpsvflfppkpkdtlmisrtpevtcvvvdvshedpevkfnw yvdgvevhnaktkpreegynstyrvvsvltvlhqdwingkeykckvsnkalgapiektiskakgqprepqvytlpps rdeltknqvsltclvkgfypsdiavewesnggpennykttppvlds dgsfflyskltvdksrwqqgnvfscsvmhealhnhytqks1s1spgggggsggggsggggsggggsevq1lesgggl vqpggslrlscaasgftfsshamswvrqapgkglewvsaiwasgegyyadsvkgrftisrdnskntlylqmnslrae dtavyycakgwlgnfdywgqgtivtvssasvaapsvfifppsdeql ksgtasvvc11nnfypreakvqwkvdnalgsgnsgesvtegdskdstyslsstltlskadyekhkvyacevthqgls spvtksfnrgec (SW11)1\1(1:307) 12. Amino acid sequences of C-terminal fusions of humanized rabbit-derived DR5-binder _________________________________________________________________________ Description Amino acid sequences SEQ
II) NO
huFc- DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
hVH007 EDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG
KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVS
LTCLVKGFYPSDIAVEWESNGQPENNYKTIPPVLDSDGSFFLYSKLTV
DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGG
SGGGGSEVQLVETGGGLIQPGGSLRLSCAASGFTVSRYAMIWVRQAPG
KGLEYIGFITSDGSTYYADSAKGRFTISRDNSKNTLYLQMNSLRAEDT
AVYYCARYTYSDGTDLWGRGTLVTVSSASTKGPSVFPLAPSSKSTSGG
TAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVV
TVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCD

hVL015 DIQMTQSPSTLSASVGDRVTITCRASQSISTYLSWYQQKPGKAPKRLI
YKASSLASGVPSRFSGSGSGTEFTLTISSLQPDDAATYYCQPNSGIAT

DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter le Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS
THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME:
NOTE POUR LE TOME / VOLUME NOTE:

Claims (42)

Claims
1. A bispecific antibody that binds to death receptor 5 (DR5) and Fibroblast Activation Protein (FAP), comprising at least one antigen binding site specific for DR5, comprising (a) a heavy chain complementarity determining region 1 (CDR1) selected from the group consisting of SEQ ID NO.:1, SEQ ID NO.:17 and SEQ ID NO.:75;
(b) a heavy chain complementarity determining region 2 (CDR2) selected from the group of SEQ ID NO.:2, SEQ ID NO.:18, SEQ ID NO.:25 and SEQ ID NO.:83;
(c) a heavy chain complementarity determining region 3 (CDR3) selected from the group of SEQ ID NO.:3, SEQ ID NO.:19, SEQ ID NO.:84, SEQ ID NO.:96, SEQ ID NO.:98;
SEQ ID NO.:104 and SEQ ID NO.:108;
(d) a light chain CDR1 selected from the group of SEQ ID NO.:4, SEQ ID NO.:20, SEQ
ID NO.:27 and SEQ ID NO.:86;
(e) a light chain CDR2 selected from the group of SEQ ID NO.:5, SEQ ID NO.:21 and SEQ ID NO.:28; and (f) a light chain CDR3 selected from the group of SEQ ID NO.:6, SEQ ID NO.:22, SEQ
ID NO.:87, SEQ ID NO.:99, SEQ ID NO.:105, SEQ ID NO.:109 and SEQ ID NO.:97;
and at least one antigen binding site specific for FAP, comprising (a) a heavy chain CDR1 selected from the group of SEQ ID NO.:9 and SEQ ID
NO.:33;
(b) a heavy chain CDR2 selected from the group of SEQ ID NO.:10 and SEQ ID
NO.:34;
(c) a heavy chain CDR3 selected from the group of SEQ ID NO.:11 and SEQ ID
NO.:35;
(d) a light chain CDR1 selected from the group of SEQ ID NO.:12 and SEQ ID
NO.:36;
(e) a light chain CDR2 selected from the group of SEQ ID NO.:13 and SEQ ID
NO.:37;
(f) a light chain CDR3 selected from the group of SEQ ID NO.:14 and SEQ ID
NO.:38.
2. The bispecific antibody of claim 1, wherein the antigen binding site specific for DR5 comprises a variable heavy chain and a variable light chain comprising an amino acid sequence selected from the group of: SEQ ID NO.:7 and SEQ ID NO.:8; SEQ ID
NO.:23 and SEQ ID NO.:24; SEQ ID NO.:26 and SEQ ID NO.:24; SEQ ID NO.:23 and SEQ ID
NO.:29; SEQ ID NO.:23 and SEQ ID NO.:30; SEQ ID NO.:26 and SEQ ID NO.:31;
SEQ ID NO.:26 and SEQ ID NO.:32; SEQ ID NO.:26 and SEQ ID NO.:30; SEQ ID
NO.:23 and SEQ ID NO.:31; SEQ ID NO.:82 and SEQ ID NO.:85; SEQ ID NO.:100 and SEQ ID NO.:101; SEQ ID NO.:102 and SEQ ID NO.:103; SEQ ID NO.:106 and SEQ ID NO.:107; SEQ ID NO.:94 and SEQ ID NO.:95;

and wherein the antigen binding site specific for FAP comprises a variable heavy chain comprising an amino acid sequence selected from the group of: SEQ ID NO.:15 and SEQ
ID NO.:39 ; and a variable light chain comprising an amino acid sequence selected from the group of SEQ ID NO.:16 and SEQ ID NO.:40.
3. The bispecific antibody of claim 1 or 2, wherein the antigen binding site specific for DR5 comprises (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO.:2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO.:4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO.:6 and the antigen binding site specific for FAP comprises (a) a heavy chain CDR1 of SEQ ID NO.:9;
(b) a heavy chain CDR2 of SEQ ID NO.:10;
(c) a heavy chain CDR3 of SEQ ID NO.:11;
(d) a light chain CDR1 of SEQ ID NO.:12;
(e) a light chain CDR2 of SEQ ID NO.:13;
(f) a light chain CDR3 of SEQ ID NO.:14.
4. The bispecific antibody of any of claims 1 to 3, wherein the antigen binding site specific for DR5 comprises a variable heavy chain comprising an amino acid sequence of SEQ ID
NO.:7 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:8;
and the antigen binding site specific for FAP comprises a heavy chain variable region comprising an amino acid sequence of SEQ ID NO.:15 and a light chain variable region comprising an amino acid sequence of SEQ ID NO.:16.
5. The bispecific antibody of any of claims 1 to 4, wherein the antibody is human.
6. The bispecific antibody of any of claims 1 to 3, wherein the antibody is humanized.
7. The bispecific antibody of any of claims 1 to 6, comprising an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DR5, and at least one Fab fragment comprising the antigen binding site specific for FAP.
8. The bispecific antibody of claim 7, wherein at least one of the Fab fragments is connected to the first or second subunit of the Fc domain via the light chain (VLCL) and at least one Fab fragment is connected to the first or second subunit of the Fc domain via the heavy chain (VHCH1).
9. The bispecific antibody of claim 7 or 8, comprising a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DR5, wherein said Fab fragments are connected at the C-terminus of the constant light chain (CL) to the first or second subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain.
10. The bispecific antibody of claim 7 or 8, comprising a) an Fc domain, b) two Fab fragments comprising an antigen binding site specific for DR5, wherein said Fab fragments are connected at the C-terminus of the constant heavy chain (CH1) to the first or second subunit of the Fc domain, c) two Fab fragments comprising the antigen binding site specific for FAP, wherein the two Fab fragments are connected at the C-terminus of the constant light chain (CL) to the first or second subunit of the Fc domain.
11. The bispecific antibody of any of claims 1 to 7, comprising an Fc domain, at least one Fab fragment comprising the antigen binding site specific for DR5, and at least one Fab fragment comprising the antigen binding site specific for FAP wherein either the variable regions or the constant regions of the heavy and light chain of at least one Fab fragment are exchanged.
12. The bispecific antibody of claim 11, comprising two Fab fragments comprising each an antigen binding site specific for DR5, and two Fab fragments comprising each an antigen binding site specific for FAP.
13. The bispecific antibody of claim 12, wherein the bispecific antibody is bivalent both for DR5 and FAP.
14. The bispecific antibody of claim 11, comprising two Fab fragments comprising each an antigen binding site specific for DR5, and one Fab fragment comprising an antigen binding site specific for FAP.
15. The bispecific antibody of claim 14, wherein the bispecific antibody is bivalent for DR5 and monovalent for FAP.
16. The bispecific antibody of claim 14, additionally comprising one Fab fragment comprising an antigen binding site specific for DR5.
17. The bispecific antibody of claim 16, wherein the bispecific antibody is trivalent for DR5 and monovalent for FAP.
18. The bispecific antibody of claim 11, comprising one Fab fragment comprising an antigen binding site specific for DR5, and one Fab fragment comprising an antigen binding site specific for FAP.
19. The bispecific antibody of claim 18, wherein the bispecific antibody is monovalent for DR5 and monovalent for FAP.
20. The bispecific antibody of any of claims 11 to 19, wherein either the variable regions or the constant regions of the heavy and light chain of the Fab fragment(s) comprising an antigen binding site specific for FAP are exchanged.
21. The bispecific antibody of any of claims 7 to 20, wherein at least one of the Fab fragments are connected to the Fc domain via a peptide linker.
22. The bispecific antibody of any of claims 7 to 21, wherein the Fc domain comprises one or more amino acid substitution that reduces binding to an Fc receptor and/or effector function.
23. The bispecific antibody of claim 22, wherein said one or more amino acid substitution is at one or more position selected from the group of L234, L235, and P329.
24. The bispecific antibody of claim 23, wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating or inhibitory Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G.
25. The bispecific antibody of any of claims 7 to 24, wherein the Fc part of the first heavy chain comprises a first dimerization module and the Fc part of the second heavy chain comprises a second dimerization module allowing a heterodimerization of the two heavy chains of the first antibody.
26. The bispecific antibody of claim 25, wherein the first dimerization module comprises knobs and the second dimerization module comprises holes according to the knobs into holes strategy.
27. An antibody that specifically binds to DR5, comprising (a) a heavy chain complementarity determining region 1 (CDR1) selected from the group consisting of SEQ ID NO.:1, SEQ ID NO.:17 and SEQ ID NO.:75;
(b) a heavy chain complementarity determining region 2(CDR2) selected from the group of SEQ ID NO.:2, SEQ ID NO.:18, SEQ ID NO.:25 and SEQ ID NO.:83;
(c) a heavy chain complementarity determining region 3 (CDR3) selected from the group of SEQ ID NO.:3, SEQ ID NO.:19, SEQ ID NO.:84, SEQ ID NO.:96, SEQ ID NO.:98, SEQ ID NO.:104 and SEQ ID NO.:108;
(d) a light chain CDR1 selected from the group of SEQ ID NO.:4, SEQ ID NO.:20, SEQ
ID NO.:27 and SEQ ID NO.:86;

(e) a light chain CDR2 selected from the group of SEQ ID NO.:5, SEQ ID NO.:21 and SEQ ID NO.:28; and (f) a light chain CDR3 selected from the group of SEQ ID NO.:6, SEQ ID NO.:22, SEQ
ID NO.:87, SEQ ID NO.:99, SEQ ID NO.:105, SEQ ID NO.:109 and SEQ ID NO.:97;
28. The antibody of claim 27, comprising (a) a heavy chain CDR1 of SEQ ID NO.:1;
(b) a heavy chain CDR2 of SEQ ID NO.:2;
(c) a heavy chain CDR3 of SEQ ID NO.:3;
(d) a light chain CDR1 of SEQ ID NO.:4;
(e) a light chain CDR2 of SEQ ID NO.:5;
(f) a light chain CDR3 of SEQ ID NO.:6.
29. The antibody of claim 27, comprising a variable heavy chain and a variable light chain comprising an amino acid sequence selected from the group of: SEQ ID NO.:7 and SEQ
ID NO.:8; SEQ ID NO.:23 and SEQ ID NO.:24; SEQ ID NO.:26 and SEQ ID NO.:24;
SEQ ID NO.:23 and SEQ ID NO.:29; SEQ ID NO.:23 and SEQ ID NO.:30; SEQ ID
NO.:26 and SEQ ID NO.:31; SEQ ID NO.:26 and SEQ ID NO.:32; SEQ ID NO.:26 and SEQ ID NO.:30; SEQ ID NO.:23 and SEQ ID NO.:31; SEQ ID NO.:82 and SEQ ID
NO.:85; SEQ ID NO.:100 and SEQ ID NO.:101; SEQ ID NO.:102 and SEQ ID
NO.:103; SEQ ID NO.:106 and SEQ ID NO.:107; SEQ ID NO.:94 and SEQ ID NO.:95.
30. The antibody of any of claims 27 to 29, comprising a variable heavy chain comprising an amino acid sequence of SEQ ID NO.:7 and a variable light chain comprising an amino acid sequence of SEQ ID NO.:8.
31. A pharmaceutical composition comprising a bispecific antibody of claims 1 to 26 or the antibody of claims 27 to 30.
32. The bispecific antibody of claims 1 to 26 or the antibody of claims 27 to 30 for the treatment of cancer.
33. The bispecific antibody of claims 1 to 26 or the antibody of claims 27 to 30 for use as a medicament.
34. Use of the bispecific antibody of claims 1 to 26 or the antibody of claims 27 to 30 in the manufacture of a medicament.
35. The use of claim 34, wherein the medicament is for treatment of cancer.
36. The use of claim 34, wherein the medicament is for treatment of pancreatic cancer or colorectal carcinoma.
37. A nucleic acid sequence comprising a sequence encoding a heavy chain of a bispecific antibody of claims 1 to 26 or the heavy chain of an antibody of claims 27 to 30.
38. A nucleic acid sequence comprising a sequence encoding a light chain of a bispecific antibody of claims 1 to 26 or the light chain of an antibody of claims 27 to 30.
39. An expression vector comprising a nucleic acid sequence of claim 37 and/or claim 38.
40. A prokaryotic or eukaryotic host cell comprising a vector according to claim 39.
41. A method of producing an antibody comprising culturing the host cell of claim 40 so that the antibody is produced.
42. The invention as described herein, especially with reference to the foregoing examples.
CA2903595A 2013-04-03 2014-04-01 Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use Expired - Fee Related CA2903595C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361808128P 2013-04-03 2013-04-03
US61/808,128 2013-04-03
PCT/EP2014/056511 WO2014161845A1 (en) 2013-04-03 2014-04-01 Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use

Publications (2)

Publication Number Publication Date
CA2903595A1 true CA2903595A1 (en) 2014-10-09
CA2903595C CA2903595C (en) 2020-03-10

Family

ID=50397172

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2903595A Expired - Fee Related CA2903595C (en) 2013-04-03 2014-04-01 Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use

Country Status (26)

Country Link
US (2) US9926379B2 (en)
EP (1) EP2981552B1 (en)
JP (2) JP6316937B2 (en)
KR (2) KR20150139909A (en)
CN (1) CN105308069B (en)
AR (1) AR095980A1 (en)
AU (1) AU2014247175B2 (en)
BR (1) BR112015025306A2 (en)
CA (1) CA2903595C (en)
CL (2) CL2015002829A1 (en)
CR (1) CR20150502A (en)
EA (1) EA201500995A1 (en)
ES (1) ES2720433T3 (en)
HK (1) HK1214604A1 (en)
IL (1) IL241795B (en)
MA (1) MA38554A3 (en)
MX (1) MX370651B (en)
PE (1) PE20160506A1 (en)
PH (1) PH12015502293A1 (en)
PL (1) PL2981552T3 (en)
SG (1) SG11201508220UA (en)
TR (1) TR201905458T4 (en)
TW (1) TWI535736B (en)
UA (1) UA118028C2 (en)
WO (1) WO2014161845A1 (en)
ZA (1) ZA201506471B (en)

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
EP2417156B1 (en) 2009-04-07 2015-02-11 Roche Glycart AG Trivalent, bispecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
CN102574921B (en) 2009-09-29 2016-05-04 罗切格利卡特公司 Bispecific death receptor agonistic antibodies
SG187746A1 (en) 2010-08-13 2013-03-28 Roche Glycart Ag Anti-fap antibodies and methods of use
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
PE20140303A1 (en) 2011-02-10 2014-03-22 Roche Glycart Ag MUTANT INTERLEUQUIN-2 POLYPEPTIDES
ES2549638T3 (en) 2011-02-28 2015-10-30 F. Hoffmann-La Roche Ag Antigen binding proteins
CN103403025B (en) 2011-02-28 2016-10-12 弗·哈夫曼-拉罗切有限公司 Monovalent antigen binding protein
KR101870555B1 (en) 2011-08-23 2018-06-22 로슈 글리카트 아게 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
RU2015100656A (en) 2012-06-27 2016-08-20 Ф. Хоффманн-Ля Рош Аг METHOD FOR PRODUCING ANTIBODY FC-FRAGMENT CONNECTING, INCLUDING AT LEAST ONE CONNECTING GROUP, WHICH SPECIALLY RELATED TO THE TARGET, AND THEIR APPLICATION
WO2014001324A1 (en) 2012-06-27 2014-01-03 Hoffmann-La Roche Ag Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US20160009824A1 (en) * 2013-03-15 2016-01-14 Merck Patent Gmbh Tetravalent bispecific antibodies
UA118028C2 (en) * 2013-04-03 2018-11-12 Рош Глікарт Аг Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use
MX2016003593A (en) 2013-10-11 2016-06-02 Hoffmann La Roche Multispecific domain exchanged common variable light chain antibodies.
JP6706578B2 (en) 2013-12-30 2020-06-10 エピムアブ バイオセラピューティクス インコーポレイテッド Tandem Fab immunoglobulins and uses thereof
AU2015254558B2 (en) * 2014-05-02 2021-02-25 Medimmune Limited Ion channel modulators and uses thereof
MX2017004526A (en) * 2014-10-08 2017-06-07 Hoffmann La Roche Combination therapy of bispecific antibodies specific for fap and dr5 and chemotherapeutic agents.
EP3224275B1 (en) 2014-11-14 2020-03-04 F.Hoffmann-La Roche Ag Antigen binding molecules comprising a tnf family ligand trimer
BR112017010324A2 (en) 2014-11-20 2018-05-15 F. Hoffmann-La Roche Ag method for treating or slowing cancer progression in an individual, molecules, methods for enhancing immune function in an individual and for selecting a patient for treatment, kits, pharmaceutical composition and uses of a combination of one molecule
MX2017006571A (en) * 2014-11-20 2017-09-29 Hoffmann La Roche T cell activating bispecific antigen binding molecules agiant folr1 and cd3.
EP3227332B1 (en) 2014-12-03 2019-11-06 F.Hoffmann-La Roche Ag Multispecific antibodies
BR112017011170A2 (en) * 2014-12-18 2018-02-27 Hoffmann La Roche method for determining the complement-dependent cytotoxicity of a composition
US20170369592A1 (en) * 2015-01-09 2017-12-28 Mabimmune Diagostics AG Novel anti-fibroblast activation protein (fap) antibodies and uses derived thereof
US10689449B2 (en) 2015-01-20 2020-06-23 Igm Biosciences, Inc. Multimeric death domain-containing receptor-5 (DR5) antibodies and uses thereof
AU2015380455A1 (en) 2015-01-26 2017-08-03 Macrogenics, Inc. Multivalent molecules comprising DR5-binding domains
WO2016156291A1 (en) 2015-03-31 2016-10-06 F. Hoffmann-La Roche Ag Antigen binding molecules comprising a trimeric tnf family ligand
WO2016168768A2 (en) 2015-04-16 2016-10-20 Alder Biopharmaceuticals, Inc. Anti-pacap antibodies and uses thereof
EP3112381A1 (en) * 2015-07-01 2017-01-04 FONDAZIONE IRCCS Istituto Nazionale dei Tumori Bispecific antibodies for use in cancer immunotherapy
CN105061604B (en) * 2015-08-19 2018-03-16 河南大学 SDR5 Fc fusion proteins mutant and its application
US20180333491A1 (en) * 2015-08-19 2018-11-22 Sinotau Pharmaceuticals Group Trail receptor-binding agents and uses of same
CN107849122B (en) * 2015-08-21 2022-01-25 豪夫迈·罗氏有限公司 Affinity chromatography purification with low conductivity wash buffer
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
BR112018002570A2 (en) 2015-10-02 2018-10-16 Hoffmann La Roche bispecific antigen binding molecule, bispecific antibody, polynucleotides, ox40-specific binding antibody, pharmaceutical composition and method for inhibiting tumor cell growth in an individual
US20180363000A1 (en) * 2015-12-14 2018-12-20 The Trustees Of The University Of Pennsylvania Aav-anti pcsk9 antibody constructs and uses thereof
CR20180365A (en) 2015-12-16 2018-09-28 Amgen Inc PROTEINS OF UNION TO THE ANTI-TL1A / ANTI-TNF-a BISPECTIVE ANTIGEN AND ITS USES
CU24613B1 (en) 2016-02-06 2022-07-08 Epimab Biotherapeutics Inc FABS TANDEM IMMUNOGLOBULIN BINDING PROTEINS (FIT-IG) BSPECIFIC BINDING TO CMET AND EGFR
FI3433280T3 (en) 2016-03-22 2023-06-06 Hoffmann La Roche Protease-activated t cell bispecific molecules
EP3231813A1 (en) * 2016-03-29 2017-10-18 F. Hoffmann-La Roche AG Trimeric costimulatory tnf family ligand-containing antigen binding molecules
US10202435B2 (en) 2016-04-15 2019-02-12 Alder Biopharmaceuticals, Inc. Anti-PACAP antibodies and uses thereof
EP3448874A4 (en) 2016-04-29 2020-04-22 Voyager Therapeutics, Inc. Compositions for the treatment of disease
EP3243836A1 (en) * 2016-05-11 2017-11-15 F. Hoffmann-La Roche AG C-terminally fused tnf family ligand trimer-containing antigen binding molecules
EP3464367B1 (en) 2016-05-27 2020-09-09 AbbVie Biotherapeutics Inc. Bispecific binding proteins binding an immunomodulatory protein and a tumor antigen
EP3472614A1 (en) * 2016-06-16 2019-04-24 F. Hoffmann-La Roche AG Assay and method for determining cdc eliciting antibodies
RU2758007C2 (en) 2016-06-30 2021-10-25 Онкорус, Инк. Delivery of therapeutic polypeptides via pseudotyped oncolytic viruses
JP2018035137A (en) 2016-07-13 2018-03-08 マブイミューン ダイアグノスティックス エイジーMabimmune Diagnostics Ag Novel anti-fibroblast activated protein (FAP) binding agent and use thereof
AU2017319318A1 (en) * 2016-08-29 2019-02-21 Tiziana Life Sciences Plc Anti-CD3 antibody formulations
JP7162592B2 (en) 2016-12-14 2022-10-28 パーデュー・リサーチ・ファウンデイション Fibroblast activation protein (FAP) targeted imaging and therapy
US11267883B2 (en) * 2016-12-21 2022-03-08 Cephalon, Inc. Antibodies that specifically bind to human IL-15 and uses thereof
EP3360898A1 (en) 2017-02-14 2018-08-15 Boehringer Ingelheim International GmbH Bispecific anti-tnf-related apoptosis-inducing ligand receptor 2 and anti-cadherin 17 binding molecules for the treatment of cancer
BR112019013189A2 (en) * 2017-01-03 2019-12-10 Hoffmann La Roche bispecific antigen binding molecules, polynucleotide, host cell, bispecific antigen binding molecule production method, pharmaceutical composition, use, methods for inhibiting tumor cell growth in an individual and for treating cancer or an infectious disease
US11613785B2 (en) 2017-01-09 2023-03-28 Onkosxcel Therapeutics, Llc Predictive and diagnostic methods for prostate cancer
BR112019017753A2 (en) * 2017-04-04 2020-04-07 Hoffmann La Roche bispecific molecule, polynucleotide, vector, cell, methods for the production of a molecule and for the treatment of an individual, composition and use of the bispecific molecule
CN108997503B (en) * 2017-06-06 2021-07-23 深圳市中科艾深医药有限公司 Human sDR5-Fc recombinant fusion protein and application thereof in preparation of medicines for treating inflammation of reproductive system
EP3710482A4 (en) * 2017-11-14 2021-08-18 University Of Virginia Patent Foundation Compositions and methods for making and using bispecific antibodies
KR102194026B1 (en) * 2017-12-15 2020-12-22 경북대학교 산학협력단 Peptides that specifically bind to TRAIL receptor and use thereof
EP3502140A1 (en) * 2017-12-21 2019-06-26 F. Hoffmann-La Roche AG Combination therapy of tumor targeted icos agonists with t-cell bispecific molecules
US11945839B2 (en) 2017-12-22 2024-04-02 Hoffmann-La Roche Inc. Depletion of light chain mispaired antibody variants by hydrophobic interaction chromatography
WO2019133847A1 (en) 2017-12-29 2019-07-04 Oncorus, Inc. Oncolytic viral delivery of therapeutic polypeptides
MY198034A (en) * 2018-12-21 2023-07-27 Hoffmann La Roche Tumor-targeted agonistic cd28 antigen binding molecules
US20220273831A1 (en) 2019-07-08 2022-09-01 3B Pharmaceuticals Gmbh Compounds comprising a fibroblast activation protein ligand and use thereof
CN114341158A (en) 2019-07-08 2022-04-12 3B制药有限公司 Compounds comprising fibroblast activation protein ligands and uses thereof
EP3763726A1 (en) 2019-07-08 2021-01-13 3B Pharmaceuticals GmbH Compounds comprising a fibroblast activation protein ligand and use thereof
JP2022540859A (en) * 2019-07-12 2022-09-20 リプム エービー Novel BSSL antibody
CN110669139A (en) * 2019-09-18 2020-01-10 沣潮医药科技(上海)有限公司 Dimeric immunoadhesins, pharmaceutical compositions and uses
US11179473B2 (en) 2020-02-21 2021-11-23 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
AU2021300362A1 (en) 2020-07-01 2023-02-23 ARS Pharmaceuticals, Inc. Anti-ASGR1 antibody conjugates and uses thereof
EP4050018A1 (en) 2021-01-07 2022-08-31 3B Pharmaceuticals GmbH Compounds comprising a fibroblast activation protein ligand and use thereof
CN116940585A (en) 2021-01-07 2023-10-24 3B制药有限公司 Compounds comprising fibroblast activation protein ligands and uses thereof
CN113336851B (en) * 2021-06-30 2021-12-24 徐州医科大学 Novel fully human anti-human B7H3 antibody, composition containing same and application thereof

Family Cites Families (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2388385B1 (en) 1977-04-18 1982-01-08 Hitachi Metals Ltd ORNAMENT FIXED BY PERMANENT MAGNETS
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5580774A (en) 1989-07-31 1996-12-03 Eli Lilly And Company Chimeric antibodies directed against a human glycoprotein antigen
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
ES2206447T3 (en) 1991-06-14 2004-05-16 Genentech, Inc. HUMANIZED ANTIBODY FOR HEREGULINE.
AU2782292A (en) 1991-09-18 1993-04-27 Sloan-Kettering Institute For Cancer Research Activated stromal fibroblast-specific antibody, f19 and methods of using the same
ES2136092T3 (en) 1991-09-23 1999-11-16 Medical Res Council PROCEDURES FOR THE PRODUCTION OF HUMANIZED ANTIBODIES.
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
GB9317423D0 (en) 1993-08-21 1993-10-06 Imp Cancer Res Tech Monoclonal antibodies
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
GB9603256D0 (en) 1996-02-16 1996-04-17 Wellcome Found Antibodies
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
DE69830315T2 (en) 1997-06-24 2006-02-02 Genentech Inc., San Francisco GALACTOSYLATED GLYCOPROTEIN CONTAINING COMPOSITIONS AND METHOD FOR THE PRODUCTION THEREOF
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
ATE419009T1 (en) 1997-10-31 2009-01-15 Genentech Inc METHODS AND COMPOSITIONS CONSISTING OF GLYCOPROTEIN GLYCOFORMS
ATE531812T1 (en) 1997-12-05 2011-11-15 Scripps Research Inst HUMANIZATION OF RODENT ANTIBODIES
AU3657899A (en) 1998-04-20 1999-11-08 James E. Bailey Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6455677B1 (en) 1998-04-30 2002-09-24 Boehringer Ingelheim International Gmbh FAPα-specific antibody with improved producibility
EP0953639A1 (en) 1998-04-30 1999-11-03 Boehringer Ingelheim International GmbH FAPalpha-specific antibody with improved producibility
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
EP2275540B1 (en) 1999-04-09 2016-03-23 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
AU782626B2 (en) 1999-10-04 2005-08-18 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
EP1229125A4 (en) 1999-10-19 2005-06-01 Kyowa Hakko Kogyo Kk Process for producing polypeptide
WO2001068708A2 (en) 2000-03-17 2001-09-20 Boehringer Ingelheim Pharma Kg Human and humanized fap-alpha-specific antibodies
ES2528794T3 (en) 2000-04-11 2015-02-12 Genentech, Inc. Multivalent antibodies and uses thereof
DE10034607A1 (en) 2000-07-20 2002-02-07 Gundram Jung Multi-specific reagent for the selective stimulation of cell surface receptors
DE10045591A1 (en) 2000-09-15 2002-04-04 Klaus Pfizenmaier Site-specific, antibody-mediated activation of proapoptotic cytokines - AMAIZe (Antibody-Mediated Apoptosis Inducing Cytokines)
US7064191B2 (en) 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
CA2424602C (en) 2000-10-06 2012-09-18 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-producing cell
US20020155109A1 (en) 2001-04-20 2002-10-24 Lynch David H. Bispecific antibodies that bind TRAIL-R1 and TRAIL-R2
NZ592087A (en) 2001-08-03 2012-11-30 Roche Glycart Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
ES2326964T3 (en) 2001-10-25 2009-10-22 Genentech, Inc. GLICOPROTEIN COMPOSITIONS.
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US7432063B2 (en) 2002-02-14 2008-10-07 Kalobios Pharmaceuticals, Inc. Methods for affinity maturation
US7691568B2 (en) 2002-04-09 2010-04-06 Kyowa Hakko Kirin Co., Ltd Antibody composition-containing medicament
ATE503829T1 (en) 2002-04-09 2011-04-15 Kyowa Hakko Kirin Co Ltd CELL WITH REDUCED OR DELETED ACTIVITY OF A PROTEIN INVOLVED IN GDP-FUCOSE TRANSPORT
AU2003236019A1 (en) 2002-04-09 2003-10-20 Kyowa Hakko Kirin Co., Ltd. Drug containing antibody composition appropriate for patient suffering from Fc Gamma RIIIa polymorphism
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
CA2481837A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
AU2003236018A1 (en) 2002-04-09 2003-10-20 Kyowa Hakko Kirin Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcGamma RECEPTOR IIIa
US7229617B2 (en) * 2002-11-27 2007-06-12 Irm Llc Methods and compositions for inducing apoptosis in cancer cells
DE60332957D1 (en) 2002-12-16 2010-07-22 Genentech Inc IMMUNOGLOBULIN VARIANTS AND ITS USES
ES2574993T3 (en) 2003-01-22 2016-06-23 Roche Glycart Ag Fusion constructs and use thereof to produce antibodies with higher Fc receptor binding affinity and effector function
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US20080241884A1 (en) 2003-10-08 2008-10-02 Kenya Shitara Fused Protein Composition
AU2004280065A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kirin Co., Ltd. Process for producing antibody composition by using RNA inhibiting the function of alpha1,6-fucosyltransferase
EA036531B1 (en) 2003-11-05 2020-11-19 Роше Гликарт Аг Type ii anti-cd20 humanized antibody (variants), pharmaceutical composition comprising these antibody variants, and use thereof
JPWO2005053742A1 (en) 2003-12-04 2007-06-28 協和醗酵工業株式会社 Medicament containing antibody composition
CN1980957A (en) 2004-03-23 2007-06-13 比奥根艾迪克Ma公司 Receptor coupling agents and therapeutic uses thereof
MXPA06011199A (en) 2004-03-31 2007-04-16 Genentech Inc Humanized anti-tgf-beta antibodies.
CA2885854C (en) 2004-04-13 2017-02-21 F. Hoffmann-La Roche Ag Anti-p-selectin antibodies
TWI380996B (en) 2004-09-17 2013-01-01 Hoffmann La Roche Anti-ox40l antibodies
EP1791565B1 (en) 2004-09-23 2016-04-20 Genentech, Inc. Cysteine engineered antibodies and conjugates
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
SG173313A1 (en) 2005-01-05 2011-08-29 Biogen Idec Inc Cripto binding molecules
US8029783B2 (en) * 2005-02-02 2011-10-04 Genentech, Inc. DR5 antibodies and articles of manufacture containing same
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
PE20071101A1 (en) 2005-08-31 2007-12-21 Amgen Inc POLYPEPTIDES AND ANTIBODIES
EP1806365A1 (en) 2006-01-05 2007-07-11 Boehringer Ingelheim International GmbH Antibody molecules specific for fibroblast activation protein and immunoconjugates containing them
KR100847010B1 (en) * 2006-07-05 2008-07-17 아주대학교산학협력단 5 5 antibody binding specifically to dr5 and composition for preventing or treating a cancer comprising the same
EP2471816A1 (en) 2006-08-30 2012-07-04 Genentech, Inc. Multispecific antibodies
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US8227577B2 (en) 2007-12-21 2012-07-24 Hoffman-La Roche Inc. Bivalent, bispecific antibodies
US8242247B2 (en) 2007-12-21 2012-08-14 Hoffmann-La Roche Inc. Bivalent, bispecific antibodies
PT2235064E (en) 2008-01-07 2016-03-01 Amgen Inc Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2010136172A1 (en) 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Tri- or tetraspecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
JP5764127B2 (en) 2009-08-17 2015-08-12 ロシュ グリクアート アーゲー Targeted immunoconjugate
CN102574921B (en) 2009-09-29 2016-05-04 罗切格利卡特公司 Bispecific death receptor agonistic antibodies
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
SG187746A1 (en) * 2010-08-13 2013-03-28 Roche Glycart Ag Anti-fap antibodies and methods of use
PE20140303A1 (en) 2011-02-10 2014-03-22 Roche Glycart Ag MUTANT INTERLEUQUIN-2 POLYPEPTIDES
PT2691417T (en) 2011-03-29 2018-10-31 Roche Glycart Ag Antibody fc variants
EA201892619A1 (en) 2011-04-29 2019-04-30 Роше Гликарт Аг IMMUNOCONJUGATES CONTAINING INTERLEUKIN-2 MUTANT POLYPETIPS
CN103748114B (en) * 2011-08-23 2017-07-21 罗切格利卡特公司 T cell activation bispecific antigen binding molecules
SI2748202T1 (en) 2011-08-23 2018-10-30 Roche Glycart Ag Bispecific antigen binding molecules
BR112015002085A2 (en) 2012-08-08 2017-12-19 Roche Glycart Ag protein, polynucleotide, vector, host cell, method for producing protein, pharmaceutical composition, protein use, method of treatment and invention
UA118028C2 (en) * 2013-04-03 2018-11-12 Рош Глікарт Аг Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use
JP6618463B2 (en) 2013-05-07 2019-12-11 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Trimeric antigen-binding molecule
WO2016156291A1 (en) 2015-03-31 2016-10-06 F. Hoffmann-La Roche Ag Antigen binding molecules comprising a trimeric tnf family ligand

Also Published As

Publication number Publication date
ZA201506471B (en) 2017-09-27
PL2981552T3 (en) 2019-07-31
EP2981552A1 (en) 2016-02-10
CL2015002829A1 (en) 2016-07-29
BR112015025306A2 (en) 2017-10-10
US11214622B2 (en) 2022-01-04
PH12015502293A1 (en) 2016-02-15
MX370651B (en) 2019-12-18
AU2014247175B2 (en) 2017-03-16
EP2981552B1 (en) 2019-02-13
PE20160506A1 (en) 2016-05-28
CN105308069B (en) 2020-09-25
JP2018046823A (en) 2018-03-29
MA38554A3 (en) 2018-12-31
JP2016515389A (en) 2016-05-30
CL2017002244A1 (en) 2018-04-13
US9926379B2 (en) 2018-03-27
UA118028C2 (en) 2018-11-12
CR20150502A (en) 2015-11-19
AU2014247175A8 (en) 2016-04-14
JP6316937B2 (en) 2018-04-25
CA2903595C (en) 2020-03-10
SG11201508220UA (en) 2015-11-27
TW201500372A (en) 2015-01-01
CN105308069A (en) 2016-02-03
MX2015013899A (en) 2016-10-21
US20190062453A1 (en) 2019-02-28
WO2014161845A1 (en) 2014-10-09
KR20150139909A (en) 2015-12-14
IL241795B (en) 2019-09-26
TR201905458T4 (en) 2019-05-21
HK1214604A1 (en) 2016-07-29
AR095980A1 (en) 2015-11-25
EA201500995A1 (en) 2016-04-29
TWI535736B (en) 2016-06-01
US20140370019A1 (en) 2014-12-18
KR20170091179A (en) 2017-08-08
ES2720433T3 (en) 2019-07-22
AU2014247175A1 (en) 2015-09-24

Similar Documents

Publication Publication Date Title
US11214622B2 (en) Antibodies specific for DR5 and methods of use
US11613587B2 (en) Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
CA3000045C (en) Anti-human cd19 antibodies with high affinity
JP2021129564A (en) Common light chains and methods of use
CN113195056A (en) Antibodies that bind to CD3
AU2015348595A1 (en) T cell activating bispecific antigen binding molecules against FOLR1 and CD3
CA2962124A1 (en) Combination therapy of bispecific antibodies specific for fap and dr5 and chemotherapeutic agents
US20210032349A1 (en) Antibodies binding to NKG2D
TW202216767A (en) Antibodies binding to cd3 and folr1

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20150902

MKLA Lapsed

Effective date: 20220301

MKLA Lapsed

Effective date: 20200831